Article Search
검색
검색 팝업 닫기

Metrics

Help

  • 1. Aims and Scope

    Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE

  • 2. Editorial Board

    Editor-in-Chief + MORE

    Editor-in-Chief
    Yong Chan Lee Professor of Medicine
    Director, Gastrointestinal Research Laboratory
    Veterans Affairs Medical Center, Univ. California San Francisco
    San Francisco, USA

    Deputy Editor

    Deputy Editor
    Jong Pil Im Seoul National University College of Medicine, Seoul, Korea
    Robert S. Bresalier University of Texas M. D. Anderson Cancer Center, Houston, USA
    Steven H. Itzkowitz Mount Sinai Medical Center, NY, USA
  • 3. Editorial Office
  • 4. Articles
  • 5. Instructions for Authors
  • 6. File Download (PDF version)
  • 7. Ethical Standards
  • 8. Peer Review

    All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
    The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.

    The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.

Search

Search

Year

to

Article Type

Online first

Split Viewer

Online first

Repurposing of Antiplatelet Agent: Cilostazol for the Treatment of Alcohol-Related Liver Disease

Jong Ryeol Eun1 , Seung Up Kim2,3

1Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Korea; 2Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea; 3Yonsei Liver Center, Severance Hospital, Seoul, Korea

Correspondence to: Seung Up Kim
ORCID https://orcid.org/0000-0002-9658-8050
E-mail ksukorea@yuhs.ac

Received: July 2, 2024; Revised: August 21, 2024; Accepted: August 21, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Gut Liver.

Published online January 8, 2025

Copyright © Gut and Liver.

Alcohol-related liver disease (ALD) is a serious global health concern, characterized by liver inflammation and progressive fibrosis. There are no Food and Drug Administration-approved drugs, thus effective treatments are needed. Severe alcoholic hepatitis (AH) is the most severe manifestation of ALD, with a 28-day mortality rate ranging from 20% to 50%. For decades, pentoxifylline, an antiplatelet agent, has been used off-label for the treatment of severe AH owing to its tumor necrosis factor-α inhibition properties. However, the STOPAH trial did not reveal the survival benefit of pentoxifylline. Consequently, pentoxifylline is no longer recommended as the first-line therapy for severe AH. In contrast, cilostazol is widely used as an antiplatelet agent in cardiovascular medicine and demonstrates promising results. Cilostazol is a selective phosphodiesterase type 3 inhibitor, whereas pentoxifylline is non-selective. Recent studies using experimental models of alcohol-induced liver injury and other liver diseases have yielded promising results. Although cilostazol shows promise for hepatoprotective effects, it has not yet been evaluated in human clinical trials. In this review, we will explore the mechanism underlying the hepatoprotective effects of cilostazol, along with the pathophysiology of alcohol-induced liver injury, addressing the pressing need for effective therapeutic options for patients with ALD.

Keywords: Alcohol-related liver disease, Antiplatelet agent, Cilostazol, Pentoxifylline, Phosphodiesterase inhibitor

Alcohol-related liver disease (ALD) is a global health problem and is associated with liver inflammation, injury, and progressive fibrosis.1 Severe alcoholic hepatitis (AH) represents the most severe form of ALD, defined by a Maddrey Discriminant Function score over 32 or a Model for End-Stage Liver Disease score exceeding 20 in patients with recent jaundice and a history of chronic alcohol use disorder.2 Despite a high 28-day mortality rate ranging from 20% to 50%, there remains a significant need for effective treatments.1-3

Pentoxifylline is a non-selective phosphodiesterase (PDE) inhibitor with vasodilating and anti-inflammatory properties.4,5 It is used in the management of peripheral vascular disease, and the U.S. Food and Drug Administration (FDA) approved its use for the symptomatic treatment of claudication.6 Additionally, pentoxifylline has been used off-label for the treatment of severe AH because it inhibits tumor necrosis factor (TNF)-α, a major cytokine in the pathogenesis of AH.7 Pentoxifylline increases the concentration of cyclic adenosine monophosphate (cAMP) by blocking membrane-bound PDE.8

Five systematic reviews were conducted to assess the efficacy of pentoxifylline for severe AH.1,9-13 One systematic review, which included several old randomized controlled trials that also involved moderate AH, reported significantly fewer hepatorenal syndrome cases among patients treated with pentoxifylline,12 while another systematic review found no effect on hepatorenal syndrome.13 Recently, the largest randomized trial, STOPAH, showed no reduction in all-cause mortality with corticosteroids or pentoxifylline at 1 month, although non-significant mortality benefit was observed with the use of steroids at 28 days.14

In recent years, several new therapies targeting the pathophysiology have been under investigation through ongoing clinical trials.15 Novel treatments aimed at addressing inflammation, oxidative stress, apoptosis, regeneration, and gut dysbiosis are being tested in AH.16-18 Cilostazol (OPC-13013), a synthetic vasodilator and antiplatelet agent, was approved in 1988 in Japan for the treatment of occlusive peripheral arterial disease.19 Thereafter, cilostazol was also approved by the FDA in 1999 for the treatment of intermittent claudication.19 Over the past 20 years, it has been widely used as a potent inhibitor of platelet aggregation and thrombosis.20 There are reports suggesting cilostazol may be more effective than pentoxifylline for intermittent claudication.21 The antiplatelet activity of cilostazol is attributed to its inhibition of PDE.20 Recent studies have identified 11 different families of PDE.22 Among these, cilostazol selectively inhibits PDE3, which is predominantly expressed in platelets, vascular smooth muscle cells, cardiac myocytes, and hepatic cells.23 In addition to its antiplatelet effect, recent studies have suggested that it has various pharmacologic effects, including anti-inflammatory, antioxidant, and antiapoptotic effects.24-26 Cilostazol has also shown beneficial effects on alcohol-induced liver injury and nonalcoholic fatty liver disease (NAFLD) in animal models.27-30 The pleiotropic effects of cilostazol are mediated by both cAMP-dependent and -independent pathways, including the AMP-activated protein kinase (AMPK) pathway.28-30 However, cilostazol has not been tested in clinical trials for treating liver diseases, particularly ALD. In this review, we will explore cilostazol based on the pathophysiology of alcohol-induced liver injury and the potential of cilostazol for the treatment of ALD.

1. TNF-α-induced apoptosis

Excessive alcohol intake increases gut permeability to bacterial endotoxin. Subsequently, lipopolysaccharide binds to the TLR4/CD14/MD2 receptor complex on Kupffer cells via portal blood flow, prompting the production of inflammatory cytokines through the MyD88-dependent or TRIF/IRF-3 pathways.31 Among these cytokines, TNF-α stands out as a potent proinflammatory cytokine primarily generated by activated macrophages (Fig. 1A). TNF-α exerts various effects mediated by TNF receptors 1 and 2, with apoptotic effects specifically mediated by TNF-R1.32 TNF-α instigates apoptosis in hepatocytes, thereby initiating liver injury, with mitochondria playing a central role in this process. Indeed, all signaling events, either directly or indirectly, converge upon the mitochondria, the executioner in TNF-α-induced apoptosis. Caspase-3, an executioner caspase, is activated following the release of cytochrome c from the mitochondria (Fig. 1B).33,34 Notably, serum TNF-α levels are significantly elevated in patients with fulminant hepatitis, and in those with AH, these levels correlate inversely with patient survival rates.35,36 Chronic ethanol consumption further escalates TNF-α production, implicating TNF-α as a promising therapeutic target for AH.32-36

Figure 1.TNF-α-induced apoptosis. (A) Gut-derived LPS binds to the TLR4/CD14/MD2 receptor complex on Kupffer cells, prompting the release of inflammatory cytokines through either the MyD88-dependent or TRIF/IRF-3 pathways. TNF-α is a major cytokine among them. (B) Apoptosis is a key consequence of cell injury and can occur through two pathways: the extrinsic pathway via death receptors and the intrinsic pathway via ER stress. The apoptotic effect of TNF-α is mediated solely by TNF-R1. The mitochondria are central to both pathways. Caspase-3, an executioner caspase, becomes activated following the release of cytochrome c from the mitochondria. TNF-α, tumor necrosis factor α; LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; CD14, cluster of differentiation 14; MD2, myeloid differentiation 2; MyD88, myeloid differentiation primary response protein 88; TRIF, TIR domain-containing adapter-inducing interferon-β; IRF-3, interferon regulatory factor 3; IL, interleukin; TRAIL, TNF-related apoptosis-inducing ligand; ER, endoplasmic reticulum; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; tBid, truncated Bid; ROS, reactive oxygen species.

2. Oxidative stress

Chronic alcohol consumption increases CYP2E1 expression, resulting in an increased acetaldehyde concentration and its accumulation in hepatocytes.37 CYP2E1, which is located in the endoplasmic reticulum and mitochondria, participates in alcohol metabolism.38 Its activation by chronic alcohol consumption leads to the formation of reactive oxygen species (ROS) (Fig. 2). ROS damage mitochondrial DNA and proteins, leading to alterations in protein molecule structures, lipid peroxidation, and DNA molecule damage.39 They further lead to the accumulation of ROS, subsequently altering macromolecules causing the occurrence and progression of existing liver damage.40 Oxidative stress is one of the important factors in ethanol-induced liver injury, and the therapeutic potential of numerous antioxidants has been explored in ALD.40-42

Figure 2.Alcohol metabolism. Chronic alcohol consumption leads to the formation of ROS. ROS cause lipid peroxidation and produce protein/DNA adducts, resulting in liver injury via direct or immune-mediated mechanisms. ROS, reactive oxygen species; ADH, alcohol dehydrogenase; ALDH, aldehyde dehydrogenase; ER, endoplasmic reticulum; CYP2E1, cytochrome P450 2E1; 4-HNE, 4-hydroxynonenal; MDA, malondialdehyde; HSC, hepatic stellate cell; TNF-α, tumor necrosis factor α; TGF-β, transforming growth factor β.

3. Cyclic adenosine monophosphate

cAMP, a key second messenger molecule, plays critical roles in metabolism, inflammation, and fibrosis in several tissues (Fig. 3A).43-48 cAMP is synthesized from ATP (adenosine triphosphate) by adenylyl cyclase (AC) in response to various signaling molecules.43 cAMP signaling is regulated by PDE, which degrades cAMP to AMP to terminate the signaling.43 Increased cAMP activates various effector molecules, including protein kinase A (PKA) and the exchange proteins activated by cAMP.43 It is clear that cAMP signaling is critical in modulating major pathogenic pathways in patients with ALD such as inflammation, steatosis, fibrosis, and even liver regeneration.43,44 Several pharmacological approaches such as the use of cAMP analogs, AC agonists, and various PDE inhibitors have been tested to study the role of cAMP signaling in both in vitro and in vivo studies.44-46 cAMP-elevating agents invariably results in decreased proinflammatory response in monocytes and Kupffer cells.44 cAMP signaling exerts beneficial effects on lipid metabolism and fibrosis.47,48 Additionally, altered cAMP signaling impairs liver regeneration.43,44 Several cAMP-elevating agents have been tested and are clinically used to treat inflammation, tissue fibrosis, asthma, and neurological disorders.49-51 In 2011 and 2014, the FDA approved two orally available PDE4-specific inhibitors, roflumilast and apremilast, respectively.49,50 These medications are used for treating severe chronic obstructive pulmonary disease, psoriasis, and psoriatic arthritis. Ibudilast is currently undergoing clinical trials.51 Regarding liver diseases, PDE inhibitors have demonstrated beneficial effects in animal models of ALD. A broad-spectrum PDE inhibitor, pentoxifylline, has been used in patients with ALD owing to its anti-inflammatory activity. Many studies have reported that cilostazol increases intracellular cAMP levels.25,28 However, despite the potential benefits of modulating cAMP signaling as an effective therapeutic strategy for ALD treatment, no human trials testing has been conducted for these inhibitors in patients with ALD.

Figure 3.cAMP and AMPK signaling pathways. (A) PDE degrades cAMP to AMP to terminate the signaling. PDE inhibitors increase cAMP levels. Increased intracellular cAMP levels attenuate inflammation, steatosis, and fibrosis, and enhance liver regeneration. (B) AMPK is activated by phosphorylation at Thr172. Upregulated AMPK activates SREBP-1 and ACC, as well as promotes hepatocyte survival. AMPK also prevents inflammation and fibrosis. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; PDE, phosphodiesterase; AMP, adenosine monophosphate; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase; ATP, adenosine triphosphate; PKA, protein kinase A; EPACs, exchange proteins activated by cAMP; HSC, hepatic stellate cell; LKB1, liver kianse B1; CaMKKβ, calcium/calmodulin-dependent protein kinase kinase beta; TNF-α, tumor necrosis factor α.

4. AMP-activated protein kinase

AMPK plays a pivotal role in controlling cellular and organism survival during metabolic stress (Fig. 3B).52 Moreover, it is crucial in determining cell survival or death in response to ROS, depending on the duration of oxidative stress exposure.52 Additionally, it is crucial for maintaining mitochondrial content and quality.53 AMPK inhibits inflammation through various mechanisms, including direct inhibition of key inflammatory proteins. Its activation by ROS can promote cell survival by inducing autophagy, mitochondrial biogenesis, and the expression of genes involved in antioxidant defense.52,53 AMPK activated fatty acid oxidation and inhibited lipogenesis in rat hepatocytes and the livers of ethanol-fed mice. Ethanol inhibits AMPK in the liver, leading to an increased activity of sterol regulatory element-binding protein 1 (SREBP-1) and acetyl-CoA carboxylase (ACC). Consequently, hepatic lipid synthesis increases while fatty acid oxidation decreases, contributing to the development of alcoholic fatty liver disease.54 Ethanol treatment, both in vitro and in vivo, has been shown to decrease hepatic AMPK activation. Experiments with AMPK activators and inhibitors demonstrated that upregulation of AMPK promotes hepatocyte survival.55 Therefore, AMPK represents an attractive therapeutic target for alcoholic fatty liver disease. Additionally, Wang et al.56 reported that the adipokine orosomucoid alleviates adipose tissue fibrosis via the AMPK pathway.

1. Attenuation of liver inflammation

The beneficial effects of cilostazol are summarized in Fig. 4. Cilostazol has been shown to decrease liver TNF-α, interleukin-1β, and nuclear factor- kappa-light chain enhancer of active B cells levels in thioacetamide-induced liver damage.57 It also decreased TNF-α levels in the common bile duct-ligated rats.58 Furthermore, cilostazol significantly suppressed lipopolysaccharide-stimulated TNF-α production in RAW264.7 murine macrophages exposed to ethanol and in the liver of binge-drinking mice.29 Additionally, cilostazol has been shown to reduce levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and to improve liver histology.57 Chronic ethanol exposure typically decreases in cAMP levels.44 Cilostazol has been demonstrated to increase intracellular cAMP levels in both in vivo and in vitro studies.25,28,59 Therefore, the anti-inflammatory effect of cilostazol was postulated to be mediated by the cAMP signaling pathway. However, Lee and Eun29 demonstrated that the anti-inflammatory effect of cilostazol is mediated by the AMPK pathway and not by the cAMP signaling pathway. This result is consistent with the findings of Park et al.26

Figure 4.Beneficial effects of cilostazol in experimental models. The effects are mediated via cAMP-dependent and -independent pathways, including the AMPK pathway. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; TNF-α, tumor necrosis factor α; IL, interleukin; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; HSC, hepatic stellate cell; α-SMA, alpha-smooth muscle actin; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase.

2. Inhibition of oxidative stress and apoptosis

Ethanol increases intracellular ROS levels in primary cultured hepatocytes.27 Oxidative stress plays an essential role in ethanol-induced liver injury.40 Cilostazol significantly inhibits ROS production in a dose-dependent manner.27 Consistently, cilostazol prevents the ethanol-induced increase in intracellular 4-hydroxynonenal levels.27 Mitochondrial permeability transition serves as a critical regulatory mechanism for cytochrome c release. Cilostazol has been shown to effectively mitigate ethanol-induced mitochondrial dysfunction in primary cultured hepatocytes, thereby preventing the release of cytochrome c from mitochondria into cytosol.27 The Bax/Bcl-2 families are important regulators of the mitochondria-dependent apoptotic pathway.33 Ethanol increases proapoptotic Bax levels but decreases antiapoptotic Bcl-2 levels.60 Cilostazol inhibits ethanol-induced increase in cleaved caspase-3 (activated caspase-3).27 These results collectively indicate that cilostazol exerts hepatoprotective effects by ameliorating oxidative stress, increasing cell viability, preserving mitochondrial function, and inhibiting apoptosis.27 The antiapoptotic effect of cilostazol was also demonstrated by Lee et al., who showed that cilostazol restores cell viability dose-dependently and inhibits apoptosis on ethanol-treated hepatocytes. Lee et al.30 suggested that the hepatoprotective effect of cilostazol is mediated by the AMPK pathway and not the cAMP signaling pathway.

3. Attenuation of fibrogenesis

Several strategies have been developed to attenuate liver fibrosis, with one important signaling pathway involved being the cAMP pathway.43,46 Increases in cAMP levels produce antifibrotic effects by inhibiting fibroblast function and extracellular matrix (ECM) protein synthesis.61 Hence, the cAMP pathway is a potential target to mitigate fibrosis. In 1999, Shimizu et al.62 first reported that OPC-13013 (cilostazol) suppresses hepatic stellate cell (HSC) activation via the cAMP signaling pathway. Cilostazol has been shown to reduce fibrogenesis in a thioacetamide-induced liver fibrosis model.63 It modulates various processes in the liver related to oxidative stress, inflammation, apoptosis, ECM, collagen deposition, as well as the cAMP pathway.63 Compared to clopidogrel, only cilostazol attenuates liver fibrosis, suggesting it may have distinct antifibrotic mechanisms in addition to its antiplatelet action.64 This finding is consistent with the results of an in vitro study, in which cilostazol attenuated HSC proliferation and the expression of α-SMA and collagen α1, indicating the direct effect of cilostazol on HSCs.59,64

4. Improvement of hepatic steatosis

The effects of aspirin, ticlopidine, and cilostazol on suppressing NAFLD have been observed, with cilostazol found to be the most effective agent.28 The authors suggested this discrepancy may be attributed to the drugs’ effects on cAMP activation. The cAMP/PKA signaling pathway plays a major role in activating the cAMP-response element-binding protein (CREB) in hepatocytes.28 In the liver, the cAMP/CREB signaling pathway regulates the expression of the key genes involved in glucose and lipid metabolism.28 Previous studies have shown that cilostazol has beneficial effects on glucose metabolism in vitro and in vivo.65 Additionally, cilostazol attenuated TNF-α-induced chronic inflammation in adipose tissue through the suppression of TNF-α production by macrophages, leading to an amelioration of systemic insulin resistance in obese diabetic mice.66 AMPK phosphorylates key metabolic enzymes and transcriptional regulators, including fatty acid synthase, SREBP-1, SREBP-2, and ACC, which are associated with controlling lipid biosynthesis.65 Cilostazol has been found to activate AMPK in vascular smooth muscle and endothelial cells.67 Furthermore, it activates AMPK and ameliorates lipid imbalances in patients with NAFLD.65 Several studies have shown promise for cilostazol in improving hepatic steatosis.68,69 Considering the common mechanisms underlying the development of hepatic steatosis, cilostazol may also be effective in alcoholic fatty liver disease.

5. Enhancement of liver regeneration and others

Cilostazol enhances liver regeneration after major hepatectomy.70,71 It increases angiogenesis-related genes, including endothelial nitric oxide synthase mRNA.72 The increased intracellular cAMP levels in the cilostazol-treated animals may have contributed to the protection of endothelial cells, improvement of the hepatic microcirculation, and preservation of hepatocellular integrity.71,72 In another study, cilostazol alleviated hepatic ischemic/reperfusion injury in rats by protecting against hepatocyte injury and improving liver function.73-75 Moreover, cilostazol improved cholestatic liver injury by significantly decreasing AST, ALT, gamma-glutamyl transpeptidase, and T-bilirubin levels.58

FDA-approved drugs for ALD are currently unavailable. For decades, pentoxifylline and glucocorticoids have been used off-label for the treatment of severe AH. Pentoxifylline is no longer recommended as a first-line treatment based on the results of the STOPAH trial. High-dose glucocorticoids remain the only medical treatment option, but they are contraindicated in cases of gastrointestinal bleeding or active infection and must be used with caution due to their various side effects.

Experimental studies on alcohol-induced or other liver injury models have demonstrated the beneficial pharmacological effects of cilostazol, including anti-inflammatory, antioxidant, antiapoptotic, and antifibrotic effects. Cilostazol has also shown beneficial effects on hepatic steatosis in an NAFLD animal model. The pleiotropic effects of cilostazol are mediated by both cAMP-dependent and -independent pathways, including the AMPK pathway. Therefore, cilostazol may be a promising candidate for ALD, particularly AH.

Despite this scientific evidence, it is unfortunate that cilostazol has yet to be tested in clinical trials. The greatest advantage of cilostazol is its established safety profile from decades of use in cardiovascular medicine. We hope that well-designed clinical studies of cilostazol, either alone or in combination with other drugs, will be conducted for ALD.

S.U.K. has served as an advisory committee member for Gilead Sciences, GSK, Bayer, Novo Nordisk, and Eisai. He is a speaker for Gilead Sciences, GSK, Bayer, Eisai, Abbive, EchoSens, MSD, Bristol-Myers Squibb, Hanhwa, Yuhan, Samil, PharmaKing, Celltrion, and Bukwang. He has also received research grants from Abbive and Bristol-Myers Squibb. And S.U.K. is an editorial board member of the journal but was not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest relevant to this article were reported.

Conceptualization; Investigation: J.R.E., S.U.K. Data curation; Visualization: J.R.E., S.U.K. Drafting of the manuscript: J.R.E. Critical revision of the manuscript for important intellectual content: J.R.E., S.U.K.

  1. Van Melkebeke L, Korf H, Tsochatzis EA, van der Merwe S, Nevens F, Verbeek J. Treatment of severe alcoholic hepatitis: a systematic review. Curr Opin Pharmacol 2021;60:91-101.
    Pubmed CrossRef
  2. Yoon EL, Kim W. Current and future treatment for alcoholic-related liver diseases. J Gastroenterol Hepatol 2023;38:1218-1226.
    Pubmed CrossRef
  3. Ramond MJ, Poynard T, Rueff B, et al. A randomized trial of prednisolone in patients with severe alcoholic hepatitis. N Engl J Med 1992;326:507-512.
    Pubmed CrossRef
  4. Aviado DM, Porter JM. Pentoxifylline: a new drug for the treatment of intermittent claudication: mechanism of action, pharmacokinetics, clinical efficacy and adverse effects. Pharmacotherapy 1984;4:297-307.
    Pubmed CrossRef
  5. Ward A, Clissold SP. Pentoxifylline: a review of its pharmacodynamic and pharmacokinetic properties, and its therapeutic efficacy. Drugs 1987;34:50-97.
    Pubmed CrossRef
  6. Baker DE, Campbell RK. Pentoxifylline: a new agent for intermittent claudication. Drug Intell Clin Pharm 1985;19:345-348.
    Pubmed CrossRef
  7. Parker R, Armstrong MJ, Corbett C, Rowe IA, Houlihan DD. Systematic review: pentoxifylline for the treatment of severe alcoholic hepatitis. Aliment Pharmacol Ther 2013;37:845-854.
    Pubmed CrossRef
  8. Jacoby D, Mohler ER 3rd. Drug treatment of intermittent claudication. Drugs 2004;64:1657-1670.
    Pubmed CrossRef
  9. Singh S, Murad MH, Chandar AK, et al. Comparative effectiveness of pharmacological interventions for severe alcoholic hepatitis: a systematic review and network meta-analysis. Gastroenterology 2015;149:958-970.
    Pubmed CrossRef
  10. Louvet A, Thursz MR, Kim DJ, et al. Corticosteroids reduce risk of death within 28 days for patients with severe alcoholic hepatitis, compared with pentoxifylline or placebo: a meta-analysis of individual data from controlled trials. Gastroenterology 2018;155:458-468.
    Pubmed CrossRef
  11. Buzzetti E, Kalafateli M, Thorburn D, et al. Pharmacological interventions for alcoholic liver disease (alcohol-related liver disease): an attempted network meta-analysis. Cochrane Database Syst Rev 2017;3:CD011646.
    Pubmed CrossRef
  12. Lee YS, Kim HJ, Kim JH, et al. Treatment of severe alcoholic hepatitis with corticosteroid, pentoxifylline, or dual therapy: a systematic review and meta-analysis. J Clin Gastroenterol 2017;51:364-377.
    Pubmed CrossRef
  13. Njei B, Do A, McCarty TR, Fortune BE. Corticosteroids versus pentoxifylline for severe alcoholic hepatitis: a sequential analysis of randomized controlled trials. J Clin Gastroenterol 2016;50:871-881.
    Pubmed KoreaMed CrossRef
  14. Thursz MR, Richardson P, Allison M, et al. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015;372:1619-1628.
    Pubmed CrossRef
  15. Philips CA, Augustine P, Yerol PK, Rajesh S, Mahadevan P. Severe alcoholic hepatitis: current perspectives. Hepat Med 2019;11:97-108.
    Pubmed KoreaMed CrossRef
  16. Singal AK, Shah VH. Current trials and novel therapeutic targets for alcoholic hepatitis. J Hepatol 2019;70:305-313.
    Pubmed CrossRef
  17. Vergis N, Atkinson SR, Thursz MR. The future of therapy for alcoholic hepatitis: beyond corticosteroids. J Hepatol 2019;70:785-787.
    Pubmed KoreaMed CrossRef
  18. Sehrawat TS, Liu M, Shah VH. The knowns and unknowns of treatment for alcoholic hepatitis. Lancet Gastroenterol Hepatol 2020;5:494-506.
    Pubmed KoreaMed CrossRef
  19. Ikeda Y. Antiplatelet therapy using cilostazol, a specific PDE3 inhibitor. Thromb Haemost 1999;82:435-438.
    Pubmed CrossRef
  20. Kumar M, Bhattacharya V. Cilostazol: a new drug in the treatment intermittent claudication. Recent Pat Cardiovasc Drug Discov 2007;2:181-185.
    Pubmed CrossRef
  21. Dawson DL, Cutler BS, Hiatt WR, et al. A comparison of cilostazol and pentoxifylline for treating intermittent claudication. Am J Med 2000;109:523-530.
    Pubmed CrossRef
  22. Conti M. Phosphodiesterases and cyclic nucleotide signaling in endocrine cells. Mol Endocrinol 2000;14:1317-1327.
    Pubmed CrossRef
  23. Jeon YH, Heo YS, Kim CM, et al. Phosphodiesterase: overview of protein structures, potential therapeutic applications and recent progress in drug development. Cell Mol Life Sci 2005;62:1198-1220.
    Pubmed KoreaMed CrossRef
  24. Lee JH, Oh GT, Park SY, et al. Cilostazol reduces atherosclerosis by inhibition of superoxide and tumor necrosis factor-alpha formation in low-density lipoprotein receptor-null mice fed high cholesterol. J Pharmacol Exp Ther 2005;313:502-509.
    Pubmed CrossRef
  25. Kim MJ, Lee JH, Park SY, et al. Protection from apoptotic cell death by cilostazol, phosphodiesterase type III inhibitor, via cAMP-dependent protein kinase activation. Pharmacol Res 2006;54:261-267.
    Pubmed CrossRef
  26. Park WS, Jung WK, Lee DY, et al. Cilostazol protects mice against endotoxin shock and attenuates LPS-induced cytokine expression in RAW 264.7 macrophages via MAPK inhibition and NF-kappaB inactivation: not involved in cAMP mechanisms. Int Immunopharmacol 2010;10:1077-1085.
    Pubmed CrossRef
  27. Xie X, Xu X, Sun C, Yu Z. Protective effects of cilostazol on ethanol-induced damage in primary cultured hepatocytes. Cell Stress Chaperones 2018;23:203-211.
    Pubmed KoreaMed CrossRef
  28. Fujita K, Nozaki Y, Wada K, et al. Effectiveness of antiplatelet drugs against experimental non-alcoholic fatty liver disease. Gut 2008;57:1583-1591.
    Pubmed CrossRef
  29. Lee YJ, Eun JR. Cilostazol decreases ethanol-mediated TNFalpha expression in RAW264.7 murine macrophage and in liver from binge drinking mice. Korean J Physiol Pharmacol 2012;16:131-138.
    Pubmed KoreaMed CrossRef
  30. Lee YJ, Shu MS, Kim JY, et al. Cilostazol protects hepatocytes against alcohol-induced apoptosis via activation of AMPK pathway. PLoS One 2019;14:e0211415.
    Pubmed KoreaMed CrossRef
  31. Gao B, Seki E, Brenner DA, et al. Innate immunity in alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 2011;300:G516-G525.
    Pubmed KoreaMed CrossRef
  32. Ding WX, Yin XM. Dissection of the multiple mechanisms of TNF-alpha-induced apoptosis in liver injury. J Cell Mol Med 2004;8:445-454.
    Pubmed KoreaMed CrossRef
  33. Malhi H, Gores GJ. Cellular and molecular mechanisms of liver injury. Gastroenterology 2008;134:1641-1654.
    Pubmed KoreaMed CrossRef
  34. Stewart S, Jones D, Day CP. Alcoholic liver disease: new insights into mechanisms and preventative strategies. Trends Mol Med 2001;7:408-413.
    Pubmed CrossRef
  35. Muto Y, Nouri-Aria KT, Meager A, Alexander GJ, Eddleston AL, Williams R. Enhanced tumour necrosis factor and interleukin-1 in fulminant hepatic failure. Lancet 1988;2:72-74.
    Pubmed CrossRef
  36. Bird GL, Sheron N, Goka AK, Alexander GJ, Williams RS. Increased plasma tumor necrosis factor in severe alcoholic hepatitis. Ann Intern Med 1990;112:917-920.
    Pubmed CrossRef
  37. Gouillon ZQ, Miyamoto K, Donohue TM, et al. Role of CYP2E1 in the pathogenesis of alcoholic liver disease: modifications by cAMP and ubiquitin-proteasome pathway. Front Biosci 1999;4:A16-A25.
    Pubmed CrossRef
  38. Dukić M, Radonjić T, Jovanović I, et al. Alcohol, inflammation, and microbiota in alcoholic liver disease. Int J Mol Sci 2023;24:3735.
    Pubmed KoreaMed CrossRef
  39. Neuman MG. Cytokines: central factors in alcoholic liver disease. Alcohol Res Health 2003;27:307-316.
    CrossRef
  40. Das SK, Vasudevan DM. Alcohol-induced oxidative stress. Life Sci 2007;81:177-187.
    Pubmed CrossRef
  41. Higuera-de la Tijera F, Servín-Caamaño AI, Cruz-Herrera J, et al. Treatment with metadoxine and its impact on early mortality in patients with severe alcoholic hepatitis. Ann Hepatol 2014;13:343-352.
    Pubmed CrossRef
  42. Nguyen-Khac E, Thevenot T, Piquet MA, et al. Glucocorticoids plus N-acetylcysteine in severe alcoholic hepatitis. N Engl J Med 2011;365:1781-1789.
    Pubmed CrossRef
  43. Wahlang B, McClain C, Barve S, Gobejishvili L. Role of cAMP and phosphodiesterase signaling in liver health and disease. Cell Signal 2018;49:105-115.
    Pubmed KoreaMed CrossRef
  44. Elnagdy M, Barve S, McClain C, Gobejishvili L. cAMP signaling in pathobiology of alcohol associated liver disease. Biomolecules 2020;10:1433.
    Pubmed KoreaMed CrossRef
  45. Lu D, Aroonsakool N, Yokoyama U, Patel HH, Insel PA. Increase in cellular cyclic AMP concentrations reverses the profibrogenic phenotype of cardiac myofibroblasts: a novel therapeutic approach for cardiac fibrosis. Mol Pharmacol 2013;84:787-793.
    Pubmed KoreaMed CrossRef
  46. El-Agroudy NN, El-Naga RN, El-Razeq RA, El-Demerdash E. Forskolin, a hedgehog signalling inhibitor, attenuates carbon tetrachloride-induced liver fibrosis in rats. Br J Pharmacol 2016;173:3248-3260.
    Pubmed KoreaMed CrossRef
  47. Avila DV, Barker DF, Zhang J, McClain CJ, Barve S, Gobejishvili L. Dysregulation of hepatic cAMP levels via altered Pde4b expression plays a critical role in alcohol-induced steatosis. J Pathol 2016;240:96-107.
    Pubmed KoreaMed CrossRef
  48. Toda K, Kumagai N, Kaneko F, et al. Pentoxifylline prevents pig serum-induced rat liver fibrosis by inhibiting interleukin-6 production. J Gastroenterol Hepatol 2009;24:860-865.
    Pubmed CrossRef
  49. DiRuggiero M, Mancuso-Stewart E, DiRuggiero D, Zirwas M. New non-steroidal topical therapies for inflammatory dermatoses: part 3: roflumilast. Skinmed 2023;21:264-268.
  50. Gupta AK, Wang T, Polla Ravi S, Bamimore MA, Piguet V, Tosti A. Systematic review of newer agents for the management of alopecia areata in adults: Janus kinase inhibitors, biologics and phosphodiesterase-4 inhibitors. J Eur Acad Dermatol Venereol 2023;37:666-679.
    Pubmed CrossRef
  51. Li MJ, Briones MS, Heinzerling KG, Kalmin MM, Shoptaw SJ. Ibudilast attenuates peripheral inflammatory effects of methamphetamine in patients with methamphetamine use disorder. Drug Alcohol Depend 2020;206:107776.
    Pubmed KoreaMed CrossRef
  52. Sid B, Verrax J, Calderon PB. Role of AMPK activation in oxidative cell damage: implications for alcohol-induced liver disease. Biochem Pharmacol 2013;86:200-209.
    Pubmed CrossRef
  53. Day EA, Ford RJ, Steinberg GR. AMPK as a therapeutic target for treating metabolic diseases. Trends Endocrinol Metab 2017;28:545-560.
    Pubmed CrossRef
  54. You M, Matsumoto M, Pacold CM, Cho WK, Crabb DW. The role of AMP-activated protein kinase in the action of ethanol in the liver. Gastroenterology 2004;127:1798-1808.
    Pubmed CrossRef
  55. Choi BK, Kim TW, Lee DR, et al. A polymethoxy flavonoids-rich Citrus aurantium extract ameliorates ethanol-induced liver injury through modulation of AMPK and Nrf2-related signals in a binge drinking mouse model. Phytother Res 2015;29:1577-1584.
    Pubmed CrossRef
  56. Wang PY, Feng JY, Zhang Z, et al. The adipokine orosomucoid alleviates adipose tissue fibrosis via the AMPK pathway. Acta Pharmacol Sin 2022;43:367-375.
    Pubmed KoreaMed CrossRef
  57. El Awdan SA, Amin MM, Hassan A. Cilostazol attenuates indices of liver damage induced by thioacetamide in albino rats through regulating inflammatory cytokines and apoptotic biomarkers. Eur J Pharmacol 2018;822:168-176.
    Pubmed CrossRef
  58. Abdel Kawy HS. Cilostazol attenuates cholestatic liver injury and its complications in common bile duct ligated rats. Eur J Pharmacol 2015;752:8-17.
    Pubmed CrossRef
  59. Han K, Zhang Y, Yang Z. Cilostazol protects rats against alcohol-induced hepatic fibrosis via suppression of TGF-β1/CTGF activation and the cAMP/Epac1 pathway. Exp Ther Med 2019;17:2381-2388.
    Pubmed KoreaMed CrossRef
  60. Cahill A, Cunningham CC, Adachi M, et al. Effects of alcohol and oxidative stress on liver pathology: the role of the mitochondrion. Alcohol Clin Exp Res 2002;26:907-915.
    Pubmed KoreaMed CrossRef
  61. Insel PA, Murray F, Yokoyama U, et al. cAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol 2012;166:447-456.
    Pubmed KoreaMed CrossRef
  62. Shimizu E, Kobayashi Y, Oki Y, Kawasaki T, Yoshimi T, Nakamura H. OPC-13013, a cyclic nucleotide phosphodiesterase type III, inhibitor, inhibits cell proliferation and transdifferentiation of cultured rat hepatic stellate cells. Life Sci 1999;64:2081-2088.
    Pubmed CrossRef
  63. El Awdan SA, Abdel Rahman RF, Ibrahim HM, et al. Regression of fibrosis by cilostazol in a rat model of thioacetamide-induced liver fibrosis: up regulation of hepatic cAMP, and modulation of inflammatory, oxidative stress and apoptotic biomarkers. PLoS One 2019;14:e0216301.
    Pubmed KoreaMed CrossRef
  64. Saito S, Hata K, Iwaisako K, et al. Cilostazol attenuates hepatic stellate cell activation and protects mice against carbon tetrachloride-induced liver fibrosis. Hepatol Res 2014;44:460-473.
    Pubmed CrossRef
  65. Oh YJ, Kim HY, Lee MH, et al. Cilostazol improves HFD-induced hepatic steatosis by upregulating hepatic STAMP2 expression through AMPK. Mol Pharmacol 2018;94:1401-1411.
    Pubmed CrossRef
  66. Wada T, Onogi Y, Kimura Y, et al. Cilostazol ameliorates systemic insulin resistance in diabetic db/db mice by suppressing chronic inflammation in adipose tissue via modulation of both adipocyte and macrophage functions. Eur J Pharmacol 2013;707:120-129.
    Pubmed CrossRef
  67. Aoki C, Hattori Y, Tomizawa A, Jojima T, Kasai K. Anti-inflammatory role of cilostazol in vascular smooth muscle cells in vitro and in vivo. J Atheroscler Thromb 2010;17:503-509.
    Pubmed CrossRef
  68. Jeon BH, Lee YH, Yun MR, et al. Increased expression of ATP-binding cassette transporter A1 (ABCA1) as a possible mechanism for the protective effect of cilostazol against hepatic steatosis. Metabolism 2015;64:1444-1453.
    Pubmed CrossRef
  69. Chen Y, Pandiri I, Joe Y, et al. Synergistic effects of cilostazol and probucol on ER stress-induced hepatic steatosis via heme oxygenase-1-dependent activation of mitochondrial biogenesis. Oxid Med Cell Longev 2016;2016:3949813.
    Pubmed KoreaMed CrossRef
  70. Ikeda Y, Matsumata T, Takenaka K, Yamagata M, Sugimachi K. Effects of doxorubicin and/or cilostazol on cancer cells during liver regeneration after two-thirds hepatectomy in rats. Oncology 1998;55:354-356.
    Pubmed CrossRef
  71. von Heesen M, Dold S, Müller S, et al. Cilostazol improves hepatic blood perfusion, microcirculation, and liver regeneration after major hepatectomy in rats. Liver Transpl 2015;21:792-800.
    Pubmed CrossRef
  72. Sugita H, Nakanuma S, Munesue S, et al. Cilostazol improves the prognosis after hepatectomy in rats with sinusoidal obstruction syndrome. J Gastroenterol Hepatol 2024;39:1413-1421.
    Pubmed CrossRef
  73. Joe Y, Zheng M, Kim HJ, et al. Cilostazol attenuates murine hepatic ischemia and reperfusion injury via heme oxygenase-dependent activation of mitochondrial biogenesis. Am J Physiol Gastrointest Liver Physiol 2015;309:G21-G29.
    Pubmed CrossRef
  74. Fujii T, Obara H, Matsubara K, et al. Oral administration of cilostazol improves survival rate after rat liver ischemia/reperfusion injury. J Surg Res 2017;213:207-214.
    Pubmed CrossRef
  75. von Heesen M, Müller S, Keppler U, et al. Preconditioning by cilostazol protects against cold hepatic ischemia-reperfusion injury. Ann Transplant 2015;20:160-168.
    Pubmed CrossRef

Article

ahead

Gut and Liver

Published online January 8, 2025

Copyright © Gut and Liver.

Repurposing of Antiplatelet Agent: Cilostazol for the Treatment of Alcohol-Related Liver Disease

Jong Ryeol Eun1 , Seung Up Kim2,3

1Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Korea; 2Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea; 3Yonsei Liver Center, Severance Hospital, Seoul, Korea

Correspondence to:Seung Up Kim
ORCID https://orcid.org/0000-0002-9658-8050
E-mail ksukorea@yuhs.ac

Received: July 2, 2024; Revised: August 21, 2024; Accepted: August 21, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Alcohol-related liver disease (ALD) is a serious global health concern, characterized by liver inflammation and progressive fibrosis. There are no Food and Drug Administration-approved drugs, thus effective treatments are needed. Severe alcoholic hepatitis (AH) is the most severe manifestation of ALD, with a 28-day mortality rate ranging from 20% to 50%. For decades, pentoxifylline, an antiplatelet agent, has been used off-label for the treatment of severe AH owing to its tumor necrosis factor-α inhibition properties. However, the STOPAH trial did not reveal the survival benefit of pentoxifylline. Consequently, pentoxifylline is no longer recommended as the first-line therapy for severe AH. In contrast, cilostazol is widely used as an antiplatelet agent in cardiovascular medicine and demonstrates promising results. Cilostazol is a selective phosphodiesterase type 3 inhibitor, whereas pentoxifylline is non-selective. Recent studies using experimental models of alcohol-induced liver injury and other liver diseases have yielded promising results. Although cilostazol shows promise for hepatoprotective effects, it has not yet been evaluated in human clinical trials. In this review, we will explore the mechanism underlying the hepatoprotective effects of cilostazol, along with the pathophysiology of alcohol-induced liver injury, addressing the pressing need for effective therapeutic options for patients with ALD.

Keywords: Alcohol-related liver disease, Antiplatelet agent, Cilostazol, Pentoxifylline, Phosphodiesterase inhibitor

INTRODUCTION

Alcohol-related liver disease (ALD) is a global health problem and is associated with liver inflammation, injury, and progressive fibrosis.1 Severe alcoholic hepatitis (AH) represents the most severe form of ALD, defined by a Maddrey Discriminant Function score over 32 or a Model for End-Stage Liver Disease score exceeding 20 in patients with recent jaundice and a history of chronic alcohol use disorder.2 Despite a high 28-day mortality rate ranging from 20% to 50%, there remains a significant need for effective treatments.1-3

Pentoxifylline is a non-selective phosphodiesterase (PDE) inhibitor with vasodilating and anti-inflammatory properties.4,5 It is used in the management of peripheral vascular disease, and the U.S. Food and Drug Administration (FDA) approved its use for the symptomatic treatment of claudication.6 Additionally, pentoxifylline has been used off-label for the treatment of severe AH because it inhibits tumor necrosis factor (TNF)-α, a major cytokine in the pathogenesis of AH.7 Pentoxifylline increases the concentration of cyclic adenosine monophosphate (cAMP) by blocking membrane-bound PDE.8

Five systematic reviews were conducted to assess the efficacy of pentoxifylline for severe AH.1,9-13 One systematic review, which included several old randomized controlled trials that also involved moderate AH, reported significantly fewer hepatorenal syndrome cases among patients treated with pentoxifylline,12 while another systematic review found no effect on hepatorenal syndrome.13 Recently, the largest randomized trial, STOPAH, showed no reduction in all-cause mortality with corticosteroids or pentoxifylline at 1 month, although non-significant mortality benefit was observed with the use of steroids at 28 days.14

In recent years, several new therapies targeting the pathophysiology have been under investigation through ongoing clinical trials.15 Novel treatments aimed at addressing inflammation, oxidative stress, apoptosis, regeneration, and gut dysbiosis are being tested in AH.16-18 Cilostazol (OPC-13013), a synthetic vasodilator and antiplatelet agent, was approved in 1988 in Japan for the treatment of occlusive peripheral arterial disease.19 Thereafter, cilostazol was also approved by the FDA in 1999 for the treatment of intermittent claudication.19 Over the past 20 years, it has been widely used as a potent inhibitor of platelet aggregation and thrombosis.20 There are reports suggesting cilostazol may be more effective than pentoxifylline for intermittent claudication.21 The antiplatelet activity of cilostazol is attributed to its inhibition of PDE.20 Recent studies have identified 11 different families of PDE.22 Among these, cilostazol selectively inhibits PDE3, which is predominantly expressed in platelets, vascular smooth muscle cells, cardiac myocytes, and hepatic cells.23 In addition to its antiplatelet effect, recent studies have suggested that it has various pharmacologic effects, including anti-inflammatory, antioxidant, and antiapoptotic effects.24-26 Cilostazol has also shown beneficial effects on alcohol-induced liver injury and nonalcoholic fatty liver disease (NAFLD) in animal models.27-30 The pleiotropic effects of cilostazol are mediated by both cAMP-dependent and -independent pathways, including the AMP-activated protein kinase (AMPK) pathway.28-30 However, cilostazol has not been tested in clinical trials for treating liver diseases, particularly ALD. In this review, we will explore cilostazol based on the pathophysiology of alcohol-induced liver injury and the potential of cilostazol for the treatment of ALD.

PATHOPHYSIOLOGY OF ALD

1. TNF-α-induced apoptosis

Excessive alcohol intake increases gut permeability to bacterial endotoxin. Subsequently, lipopolysaccharide binds to the TLR4/CD14/MD2 receptor complex on Kupffer cells via portal blood flow, prompting the production of inflammatory cytokines through the MyD88-dependent or TRIF/IRF-3 pathways.31 Among these cytokines, TNF-α stands out as a potent proinflammatory cytokine primarily generated by activated macrophages (Fig. 1A). TNF-α exerts various effects mediated by TNF receptors 1 and 2, with apoptotic effects specifically mediated by TNF-R1.32 TNF-α instigates apoptosis in hepatocytes, thereby initiating liver injury, with mitochondria playing a central role in this process. Indeed, all signaling events, either directly or indirectly, converge upon the mitochondria, the executioner in TNF-α-induced apoptosis. Caspase-3, an executioner caspase, is activated following the release of cytochrome c from the mitochondria (Fig. 1B).33,34 Notably, serum TNF-α levels are significantly elevated in patients with fulminant hepatitis, and in those with AH, these levels correlate inversely with patient survival rates.35,36 Chronic ethanol consumption further escalates TNF-α production, implicating TNF-α as a promising therapeutic target for AH.32-36

Figure 1. TNF-α-induced apoptosis. (A) Gut-derived LPS binds to the TLR4/CD14/MD2 receptor complex on Kupffer cells, prompting the release of inflammatory cytokines through either the MyD88-dependent or TRIF/IRF-3 pathways. TNF-α is a major cytokine among them. (B) Apoptosis is a key consequence of cell injury and can occur through two pathways: the extrinsic pathway via death receptors and the intrinsic pathway via ER stress. The apoptotic effect of TNF-α is mediated solely by TNF-R1. The mitochondria are central to both pathways. Caspase-3, an executioner caspase, becomes activated following the release of cytochrome c from the mitochondria. TNF-α, tumor necrosis factor α; LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; CD14, cluster of differentiation 14; MD2, myeloid differentiation 2; MyD88, myeloid differentiation primary response protein 88; TRIF, TIR domain-containing adapter-inducing interferon-β; IRF-3, interferon regulatory factor 3; IL, interleukin; TRAIL, TNF-related apoptosis-inducing ligand; ER, endoplasmic reticulum; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; tBid, truncated Bid; ROS, reactive oxygen species.

2. Oxidative stress

Chronic alcohol consumption increases CYP2E1 expression, resulting in an increased acetaldehyde concentration and its accumulation in hepatocytes.37 CYP2E1, which is located in the endoplasmic reticulum and mitochondria, participates in alcohol metabolism.38 Its activation by chronic alcohol consumption leads to the formation of reactive oxygen species (ROS) (Fig. 2). ROS damage mitochondrial DNA and proteins, leading to alterations in protein molecule structures, lipid peroxidation, and DNA molecule damage.39 They further lead to the accumulation of ROS, subsequently altering macromolecules causing the occurrence and progression of existing liver damage.40 Oxidative stress is one of the important factors in ethanol-induced liver injury, and the therapeutic potential of numerous antioxidants has been explored in ALD.40-42

Figure 2. Alcohol metabolism. Chronic alcohol consumption leads to the formation of ROS. ROS cause lipid peroxidation and produce protein/DNA adducts, resulting in liver injury via direct or immune-mediated mechanisms. ROS, reactive oxygen species; ADH, alcohol dehydrogenase; ALDH, aldehyde dehydrogenase; ER, endoplasmic reticulum; CYP2E1, cytochrome P450 2E1; 4-HNE, 4-hydroxynonenal; MDA, malondialdehyde; HSC, hepatic stellate cell; TNF-α, tumor necrosis factor α; TGF-β, transforming growth factor β.

3. Cyclic adenosine monophosphate

cAMP, a key second messenger molecule, plays critical roles in metabolism, inflammation, and fibrosis in several tissues (Fig. 3A).43-48 cAMP is synthesized from ATP (adenosine triphosphate) by adenylyl cyclase (AC) in response to various signaling molecules.43 cAMP signaling is regulated by PDE, which degrades cAMP to AMP to terminate the signaling.43 Increased cAMP activates various effector molecules, including protein kinase A (PKA) and the exchange proteins activated by cAMP.43 It is clear that cAMP signaling is critical in modulating major pathogenic pathways in patients with ALD such as inflammation, steatosis, fibrosis, and even liver regeneration.43,44 Several pharmacological approaches such as the use of cAMP analogs, AC agonists, and various PDE inhibitors have been tested to study the role of cAMP signaling in both in vitro and in vivo studies.44-46 cAMP-elevating agents invariably results in decreased proinflammatory response in monocytes and Kupffer cells.44 cAMP signaling exerts beneficial effects on lipid metabolism and fibrosis.47,48 Additionally, altered cAMP signaling impairs liver regeneration.43,44 Several cAMP-elevating agents have been tested and are clinically used to treat inflammation, tissue fibrosis, asthma, and neurological disorders.49-51 In 2011 and 2014, the FDA approved two orally available PDE4-specific inhibitors, roflumilast and apremilast, respectively.49,50 These medications are used for treating severe chronic obstructive pulmonary disease, psoriasis, and psoriatic arthritis. Ibudilast is currently undergoing clinical trials.51 Regarding liver diseases, PDE inhibitors have demonstrated beneficial effects in animal models of ALD. A broad-spectrum PDE inhibitor, pentoxifylline, has been used in patients with ALD owing to its anti-inflammatory activity. Many studies have reported that cilostazol increases intracellular cAMP levels.25,28 However, despite the potential benefits of modulating cAMP signaling as an effective therapeutic strategy for ALD treatment, no human trials testing has been conducted for these inhibitors in patients with ALD.

Figure 3. cAMP and AMPK signaling pathways. (A) PDE degrades cAMP to AMP to terminate the signaling. PDE inhibitors increase cAMP levels. Increased intracellular cAMP levels attenuate inflammation, steatosis, and fibrosis, and enhance liver regeneration. (B) AMPK is activated by phosphorylation at Thr172. Upregulated AMPK activates SREBP-1 and ACC, as well as promotes hepatocyte survival. AMPK also prevents inflammation and fibrosis. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; PDE, phosphodiesterase; AMP, adenosine monophosphate; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase; ATP, adenosine triphosphate; PKA, protein kinase A; EPACs, exchange proteins activated by cAMP; HSC, hepatic stellate cell; LKB1, liver kianse B1; CaMKKβ, calcium/calmodulin-dependent protein kinase kinase beta; TNF-α, tumor necrosis factor α.

4. AMP-activated protein kinase

AMPK plays a pivotal role in controlling cellular and organism survival during metabolic stress (Fig. 3B).52 Moreover, it is crucial in determining cell survival or death in response to ROS, depending on the duration of oxidative stress exposure.52 Additionally, it is crucial for maintaining mitochondrial content and quality.53 AMPK inhibits inflammation through various mechanisms, including direct inhibition of key inflammatory proteins. Its activation by ROS can promote cell survival by inducing autophagy, mitochondrial biogenesis, and the expression of genes involved in antioxidant defense.52,53 AMPK activated fatty acid oxidation and inhibited lipogenesis in rat hepatocytes and the livers of ethanol-fed mice. Ethanol inhibits AMPK in the liver, leading to an increased activity of sterol regulatory element-binding protein 1 (SREBP-1) and acetyl-CoA carboxylase (ACC). Consequently, hepatic lipid synthesis increases while fatty acid oxidation decreases, contributing to the development of alcoholic fatty liver disease.54 Ethanol treatment, both in vitro and in vivo, has been shown to decrease hepatic AMPK activation. Experiments with AMPK activators and inhibitors demonstrated that upregulation of AMPK promotes hepatocyte survival.55 Therefore, AMPK represents an attractive therapeutic target for alcoholic fatty liver disease. Additionally, Wang et al.56 reported that the adipokine orosomucoid alleviates adipose tissue fibrosis via the AMPK pathway.

BENEFICIAL EFFECTS OF CILOSTAZOL ON LIVER INJURY MODELS

1. Attenuation of liver inflammation

The beneficial effects of cilostazol are summarized in Fig. 4. Cilostazol has been shown to decrease liver TNF-α, interleukin-1β, and nuclear factor- kappa-light chain enhancer of active B cells levels in thioacetamide-induced liver damage.57 It also decreased TNF-α levels in the common bile duct-ligated rats.58 Furthermore, cilostazol significantly suppressed lipopolysaccharide-stimulated TNF-α production in RAW264.7 murine macrophages exposed to ethanol and in the liver of binge-drinking mice.29 Additionally, cilostazol has been shown to reduce levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and to improve liver histology.57 Chronic ethanol exposure typically decreases in cAMP levels.44 Cilostazol has been demonstrated to increase intracellular cAMP levels in both in vivo and in vitro studies.25,28,59 Therefore, the anti-inflammatory effect of cilostazol was postulated to be mediated by the cAMP signaling pathway. However, Lee and Eun29 demonstrated that the anti-inflammatory effect of cilostazol is mediated by the AMPK pathway and not by the cAMP signaling pathway. This result is consistent with the findings of Park et al.26

Figure 4. Beneficial effects of cilostazol in experimental models. The effects are mediated via cAMP-dependent and -independent pathways, including the AMPK pathway. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; TNF-α, tumor necrosis factor α; IL, interleukin; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; HSC, hepatic stellate cell; α-SMA, alpha-smooth muscle actin; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase.

2. Inhibition of oxidative stress and apoptosis

Ethanol increases intracellular ROS levels in primary cultured hepatocytes.27 Oxidative stress plays an essential role in ethanol-induced liver injury.40 Cilostazol significantly inhibits ROS production in a dose-dependent manner.27 Consistently, cilostazol prevents the ethanol-induced increase in intracellular 4-hydroxynonenal levels.27 Mitochondrial permeability transition serves as a critical regulatory mechanism for cytochrome c release. Cilostazol has been shown to effectively mitigate ethanol-induced mitochondrial dysfunction in primary cultured hepatocytes, thereby preventing the release of cytochrome c from mitochondria into cytosol.27 The Bax/Bcl-2 families are important regulators of the mitochondria-dependent apoptotic pathway.33 Ethanol increases proapoptotic Bax levels but decreases antiapoptotic Bcl-2 levels.60 Cilostazol inhibits ethanol-induced increase in cleaved caspase-3 (activated caspase-3).27 These results collectively indicate that cilostazol exerts hepatoprotective effects by ameliorating oxidative stress, increasing cell viability, preserving mitochondrial function, and inhibiting apoptosis.27 The antiapoptotic effect of cilostazol was also demonstrated by Lee et al., who showed that cilostazol restores cell viability dose-dependently and inhibits apoptosis on ethanol-treated hepatocytes. Lee et al.30 suggested that the hepatoprotective effect of cilostazol is mediated by the AMPK pathway and not the cAMP signaling pathway.

3. Attenuation of fibrogenesis

Several strategies have been developed to attenuate liver fibrosis, with one important signaling pathway involved being the cAMP pathway.43,46 Increases in cAMP levels produce antifibrotic effects by inhibiting fibroblast function and extracellular matrix (ECM) protein synthesis.61 Hence, the cAMP pathway is a potential target to mitigate fibrosis. In 1999, Shimizu et al.62 first reported that OPC-13013 (cilostazol) suppresses hepatic stellate cell (HSC) activation via the cAMP signaling pathway. Cilostazol has been shown to reduce fibrogenesis in a thioacetamide-induced liver fibrosis model.63 It modulates various processes in the liver related to oxidative stress, inflammation, apoptosis, ECM, collagen deposition, as well as the cAMP pathway.63 Compared to clopidogrel, only cilostazol attenuates liver fibrosis, suggesting it may have distinct antifibrotic mechanisms in addition to its antiplatelet action.64 This finding is consistent with the results of an in vitro study, in which cilostazol attenuated HSC proliferation and the expression of α-SMA and collagen α1, indicating the direct effect of cilostazol on HSCs.59,64

4. Improvement of hepatic steatosis

The effects of aspirin, ticlopidine, and cilostazol on suppressing NAFLD have been observed, with cilostazol found to be the most effective agent.28 The authors suggested this discrepancy may be attributed to the drugs’ effects on cAMP activation. The cAMP/PKA signaling pathway plays a major role in activating the cAMP-response element-binding protein (CREB) in hepatocytes.28 In the liver, the cAMP/CREB signaling pathway regulates the expression of the key genes involved in glucose and lipid metabolism.28 Previous studies have shown that cilostazol has beneficial effects on glucose metabolism in vitro and in vivo.65 Additionally, cilostazol attenuated TNF-α-induced chronic inflammation in adipose tissue through the suppression of TNF-α production by macrophages, leading to an amelioration of systemic insulin resistance in obese diabetic mice.66 AMPK phosphorylates key metabolic enzymes and transcriptional regulators, including fatty acid synthase, SREBP-1, SREBP-2, and ACC, which are associated with controlling lipid biosynthesis.65 Cilostazol has been found to activate AMPK in vascular smooth muscle and endothelial cells.67 Furthermore, it activates AMPK and ameliorates lipid imbalances in patients with NAFLD.65 Several studies have shown promise for cilostazol in improving hepatic steatosis.68,69 Considering the common mechanisms underlying the development of hepatic steatosis, cilostazol may also be effective in alcoholic fatty liver disease.

5. Enhancement of liver regeneration and others

Cilostazol enhances liver regeneration after major hepatectomy.70,71 It increases angiogenesis-related genes, including endothelial nitric oxide synthase mRNA.72 The increased intracellular cAMP levels in the cilostazol-treated animals may have contributed to the protection of endothelial cells, improvement of the hepatic microcirculation, and preservation of hepatocellular integrity.71,72 In another study, cilostazol alleviated hepatic ischemic/reperfusion injury in rats by protecting against hepatocyte injury and improving liver function.73-75 Moreover, cilostazol improved cholestatic liver injury by significantly decreasing AST, ALT, gamma-glutamyl transpeptidase, and T-bilirubin levels.58

CONCLUSION

FDA-approved drugs for ALD are currently unavailable. For decades, pentoxifylline and glucocorticoids have been used off-label for the treatment of severe AH. Pentoxifylline is no longer recommended as a first-line treatment based on the results of the STOPAH trial. High-dose glucocorticoids remain the only medical treatment option, but they are contraindicated in cases of gastrointestinal bleeding or active infection and must be used with caution due to their various side effects.

Experimental studies on alcohol-induced or other liver injury models have demonstrated the beneficial pharmacological effects of cilostazol, including anti-inflammatory, antioxidant, antiapoptotic, and antifibrotic effects. Cilostazol has also shown beneficial effects on hepatic steatosis in an NAFLD animal model. The pleiotropic effects of cilostazol are mediated by both cAMP-dependent and -independent pathways, including the AMPK pathway. Therefore, cilostazol may be a promising candidate for ALD, particularly AH.

Despite this scientific evidence, it is unfortunate that cilostazol has yet to be tested in clinical trials. The greatest advantage of cilostazol is its established safety profile from decades of use in cardiovascular medicine. We hope that well-designed clinical studies of cilostazol, either alone or in combination with other drugs, will be conducted for ALD.

CONFLICTS OF INTEREST

S.U.K. has served as an advisory committee member for Gilead Sciences, GSK, Bayer, Novo Nordisk, and Eisai. He is a speaker for Gilead Sciences, GSK, Bayer, Eisai, Abbive, EchoSens, MSD, Bristol-Myers Squibb, Hanhwa, Yuhan, Samil, PharmaKing, Celltrion, and Bukwang. He has also received research grants from Abbive and Bristol-Myers Squibb. And S.U.K. is an editorial board member of the journal but was not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest relevant to this article were reported.

AUTHOR CONTRIBUTIONS

Conceptualization; Investigation: J.R.E., S.U.K. Data curation; Visualization: J.R.E., S.U.K. Drafting of the manuscript: J.R.E. Critical revision of the manuscript for important intellectual content: J.R.E., S.U.K.

Fig 1.

Figure 1.TNF-α-induced apoptosis. (A) Gut-derived LPS binds to the TLR4/CD14/MD2 receptor complex on Kupffer cells, prompting the release of inflammatory cytokines through either the MyD88-dependent or TRIF/IRF-3 pathways. TNF-α is a major cytokine among them. (B) Apoptosis is a key consequence of cell injury and can occur through two pathways: the extrinsic pathway via death receptors and the intrinsic pathway via ER stress. The apoptotic effect of TNF-α is mediated solely by TNF-R1. The mitochondria are central to both pathways. Caspase-3, an executioner caspase, becomes activated following the release of cytochrome c from the mitochondria. TNF-α, tumor necrosis factor α; LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; CD14, cluster of differentiation 14; MD2, myeloid differentiation 2; MyD88, myeloid differentiation primary response protein 88; TRIF, TIR domain-containing adapter-inducing interferon-β; IRF-3, interferon regulatory factor 3; IL, interleukin; TRAIL, TNF-related apoptosis-inducing ligand; ER, endoplasmic reticulum; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; tBid, truncated Bid; ROS, reactive oxygen species.
Gut and Liver 2025; :

Fig 2.

Figure 2.Alcohol metabolism. Chronic alcohol consumption leads to the formation of ROS. ROS cause lipid peroxidation and produce protein/DNA adducts, resulting in liver injury via direct or immune-mediated mechanisms. ROS, reactive oxygen species; ADH, alcohol dehydrogenase; ALDH, aldehyde dehydrogenase; ER, endoplasmic reticulum; CYP2E1, cytochrome P450 2E1; 4-HNE, 4-hydroxynonenal; MDA, malondialdehyde; HSC, hepatic stellate cell; TNF-α, tumor necrosis factor α; TGF-β, transforming growth factor β.
Gut and Liver 2025; :

Fig 3.

Figure 3.cAMP and AMPK signaling pathways. (A) PDE degrades cAMP to AMP to terminate the signaling. PDE inhibitors increase cAMP levels. Increased intracellular cAMP levels attenuate inflammation, steatosis, and fibrosis, and enhance liver regeneration. (B) AMPK is activated by phosphorylation at Thr172. Upregulated AMPK activates SREBP-1 and ACC, as well as promotes hepatocyte survival. AMPK also prevents inflammation and fibrosis. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; PDE, phosphodiesterase; AMP, adenosine monophosphate; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase; ATP, adenosine triphosphate; PKA, protein kinase A; EPACs, exchange proteins activated by cAMP; HSC, hepatic stellate cell; LKB1, liver kianse B1; CaMKKβ, calcium/calmodulin-dependent protein kinase kinase beta; TNF-α, tumor necrosis factor α.
Gut and Liver 2025; :

Fig 4.

Figure 4.Beneficial effects of cilostazol in experimental models. The effects are mediated via cAMP-dependent and -independent pathways, including the AMPK pathway. cAMP, cyclic adenosine monophosphate; AMPK, adenosine monophosphate-activated protein kinase; TNF-α, tumor necrosis factor α; IL, interleukin; NF-κB, nuclear factor-kappa-light chain enhancer of active B cells; HSC, hepatic stellate cell; α-SMA, alpha-smooth muscle actin; SREBP-1, sterol regulatory element-binding protein 1; ACC, acetyl-CoA carboxylase.
Gut and Liver 2025; :

References

  1. Van Melkebeke L, Korf H, Tsochatzis EA, van der Merwe S, Nevens F, Verbeek J. Treatment of severe alcoholic hepatitis: a systematic review. Curr Opin Pharmacol 2021;60:91-101.
    Pubmed CrossRef
  2. Yoon EL, Kim W. Current and future treatment for alcoholic-related liver diseases. J Gastroenterol Hepatol 2023;38:1218-1226.
    Pubmed CrossRef
  3. Ramond MJ, Poynard T, Rueff B, et al. A randomized trial of prednisolone in patients with severe alcoholic hepatitis. N Engl J Med 1992;326:507-512.
    Pubmed CrossRef
  4. Aviado DM, Porter JM. Pentoxifylline: a new drug for the treatment of intermittent claudication: mechanism of action, pharmacokinetics, clinical efficacy and adverse effects. Pharmacotherapy 1984;4:297-307.
    Pubmed CrossRef
  5. Ward A, Clissold SP. Pentoxifylline: a review of its pharmacodynamic and pharmacokinetic properties, and its therapeutic efficacy. Drugs 1987;34:50-97.
    Pubmed CrossRef
  6. Baker DE, Campbell RK. Pentoxifylline: a new agent for intermittent claudication. Drug Intell Clin Pharm 1985;19:345-348.
    Pubmed CrossRef
  7. Parker R, Armstrong MJ, Corbett C, Rowe IA, Houlihan DD. Systematic review: pentoxifylline for the treatment of severe alcoholic hepatitis. Aliment Pharmacol Ther 2013;37:845-854.
    Pubmed CrossRef
  8. Jacoby D, Mohler ER 3rd. Drug treatment of intermittent claudication. Drugs 2004;64:1657-1670.
    Pubmed CrossRef
  9. Singh S, Murad MH, Chandar AK, et al. Comparative effectiveness of pharmacological interventions for severe alcoholic hepatitis: a systematic review and network meta-analysis. Gastroenterology 2015;149:958-970.
    Pubmed CrossRef
  10. Louvet A, Thursz MR, Kim DJ, et al. Corticosteroids reduce risk of death within 28 days for patients with severe alcoholic hepatitis, compared with pentoxifylline or placebo: a meta-analysis of individual data from controlled trials. Gastroenterology 2018;155:458-468.
    Pubmed CrossRef
  11. Buzzetti E, Kalafateli M, Thorburn D, et al. Pharmacological interventions for alcoholic liver disease (alcohol-related liver disease): an attempted network meta-analysis. Cochrane Database Syst Rev 2017;3:CD011646.
    Pubmed CrossRef
  12. Lee YS, Kim HJ, Kim JH, et al. Treatment of severe alcoholic hepatitis with corticosteroid, pentoxifylline, or dual therapy: a systematic review and meta-analysis. J Clin Gastroenterol 2017;51:364-377.
    Pubmed CrossRef
  13. Njei B, Do A, McCarty TR, Fortune BE. Corticosteroids versus pentoxifylline for severe alcoholic hepatitis: a sequential analysis of randomized controlled trials. J Clin Gastroenterol 2016;50:871-881.
    Pubmed KoreaMed CrossRef
  14. Thursz MR, Richardson P, Allison M, et al. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015;372:1619-1628.
    Pubmed CrossRef
  15. Philips CA, Augustine P, Yerol PK, Rajesh S, Mahadevan P. Severe alcoholic hepatitis: current perspectives. Hepat Med 2019;11:97-108.
    Pubmed KoreaMed CrossRef
  16. Singal AK, Shah VH. Current trials and novel therapeutic targets for alcoholic hepatitis. J Hepatol 2019;70:305-313.
    Pubmed CrossRef
  17. Vergis N, Atkinson SR, Thursz MR. The future of therapy for alcoholic hepatitis: beyond corticosteroids. J Hepatol 2019;70:785-787.
    Pubmed KoreaMed CrossRef
  18. Sehrawat TS, Liu M, Shah VH. The knowns and unknowns of treatment for alcoholic hepatitis. Lancet Gastroenterol Hepatol 2020;5:494-506.
    Pubmed KoreaMed CrossRef
  19. Ikeda Y. Antiplatelet therapy using cilostazol, a specific PDE3 inhibitor. Thromb Haemost 1999;82:435-438.
    Pubmed CrossRef
  20. Kumar M, Bhattacharya V. Cilostazol: a new drug in the treatment intermittent claudication. Recent Pat Cardiovasc Drug Discov 2007;2:181-185.
    Pubmed CrossRef
  21. Dawson DL, Cutler BS, Hiatt WR, et al. A comparison of cilostazol and pentoxifylline for treating intermittent claudication. Am J Med 2000;109:523-530.
    Pubmed CrossRef
  22. Conti M. Phosphodiesterases and cyclic nucleotide signaling in endocrine cells. Mol Endocrinol 2000;14:1317-1327.
    Pubmed CrossRef
  23. Jeon YH, Heo YS, Kim CM, et al. Phosphodiesterase: overview of protein structures, potential therapeutic applications and recent progress in drug development. Cell Mol Life Sci 2005;62:1198-1220.
    Pubmed KoreaMed CrossRef
  24. Lee JH, Oh GT, Park SY, et al. Cilostazol reduces atherosclerosis by inhibition of superoxide and tumor necrosis factor-alpha formation in low-density lipoprotein receptor-null mice fed high cholesterol. J Pharmacol Exp Ther 2005;313:502-509.
    Pubmed CrossRef
  25. Kim MJ, Lee JH, Park SY, et al. Protection from apoptotic cell death by cilostazol, phosphodiesterase type III inhibitor, via cAMP-dependent protein kinase activation. Pharmacol Res 2006;54:261-267.
    Pubmed CrossRef
  26. Park WS, Jung WK, Lee DY, et al. Cilostazol protects mice against endotoxin shock and attenuates LPS-induced cytokine expression in RAW 264.7 macrophages via MAPK inhibition and NF-kappaB inactivation: not involved in cAMP mechanisms. Int Immunopharmacol 2010;10:1077-1085.
    Pubmed CrossRef
  27. Xie X, Xu X, Sun C, Yu Z. Protective effects of cilostazol on ethanol-induced damage in primary cultured hepatocytes. Cell Stress Chaperones 2018;23:203-211.
    Pubmed KoreaMed CrossRef
  28. Fujita K, Nozaki Y, Wada K, et al. Effectiveness of antiplatelet drugs against experimental non-alcoholic fatty liver disease. Gut 2008;57:1583-1591.
    Pubmed CrossRef
  29. Lee YJ, Eun JR. Cilostazol decreases ethanol-mediated TNFalpha expression in RAW264.7 murine macrophage and in liver from binge drinking mice. Korean J Physiol Pharmacol 2012;16:131-138.
    Pubmed KoreaMed CrossRef
  30. Lee YJ, Shu MS, Kim JY, et al. Cilostazol protects hepatocytes against alcohol-induced apoptosis via activation of AMPK pathway. PLoS One 2019;14:e0211415.
    Pubmed KoreaMed CrossRef
  31. Gao B, Seki E, Brenner DA, et al. Innate immunity in alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 2011;300:G516-G525.
    Pubmed KoreaMed CrossRef
  32. Ding WX, Yin XM. Dissection of the multiple mechanisms of TNF-alpha-induced apoptosis in liver injury. J Cell Mol Med 2004;8:445-454.
    Pubmed KoreaMed CrossRef
  33. Malhi H, Gores GJ. Cellular and molecular mechanisms of liver injury. Gastroenterology 2008;134:1641-1654.
    Pubmed KoreaMed CrossRef
  34. Stewart S, Jones D, Day CP. Alcoholic liver disease: new insights into mechanisms and preventative strategies. Trends Mol Med 2001;7:408-413.
    Pubmed CrossRef
  35. Muto Y, Nouri-Aria KT, Meager A, Alexander GJ, Eddleston AL, Williams R. Enhanced tumour necrosis factor and interleukin-1 in fulminant hepatic failure. Lancet 1988;2:72-74.
    Pubmed CrossRef
  36. Bird GL, Sheron N, Goka AK, Alexander GJ, Williams RS. Increased plasma tumor necrosis factor in severe alcoholic hepatitis. Ann Intern Med 1990;112:917-920.
    Pubmed CrossRef
  37. Gouillon ZQ, Miyamoto K, Donohue TM, et al. Role of CYP2E1 in the pathogenesis of alcoholic liver disease: modifications by cAMP and ubiquitin-proteasome pathway. Front Biosci 1999;4:A16-A25.
    Pubmed CrossRef
  38. Dukić M, Radonjić T, Jovanović I, et al. Alcohol, inflammation, and microbiota in alcoholic liver disease. Int J Mol Sci 2023;24:3735.
    Pubmed KoreaMed CrossRef
  39. Neuman MG. Cytokines: central factors in alcoholic liver disease. Alcohol Res Health 2003;27:307-316.
    CrossRef
  40. Das SK, Vasudevan DM. Alcohol-induced oxidative stress. Life Sci 2007;81:177-187.
    Pubmed CrossRef
  41. Higuera-de la Tijera F, Servín-Caamaño AI, Cruz-Herrera J, et al. Treatment with metadoxine and its impact on early mortality in patients with severe alcoholic hepatitis. Ann Hepatol 2014;13:343-352.
    Pubmed CrossRef
  42. Nguyen-Khac E, Thevenot T, Piquet MA, et al. Glucocorticoids plus N-acetylcysteine in severe alcoholic hepatitis. N Engl J Med 2011;365:1781-1789.
    Pubmed CrossRef
  43. Wahlang B, McClain C, Barve S, Gobejishvili L. Role of cAMP and phosphodiesterase signaling in liver health and disease. Cell Signal 2018;49:105-115.
    Pubmed KoreaMed CrossRef
  44. Elnagdy M, Barve S, McClain C, Gobejishvili L. cAMP signaling in pathobiology of alcohol associated liver disease. Biomolecules 2020;10:1433.
    Pubmed KoreaMed CrossRef
  45. Lu D, Aroonsakool N, Yokoyama U, Patel HH, Insel PA. Increase in cellular cyclic AMP concentrations reverses the profibrogenic phenotype of cardiac myofibroblasts: a novel therapeutic approach for cardiac fibrosis. Mol Pharmacol 2013;84:787-793.
    Pubmed KoreaMed CrossRef
  46. El-Agroudy NN, El-Naga RN, El-Razeq RA, El-Demerdash E. Forskolin, a hedgehog signalling inhibitor, attenuates carbon tetrachloride-induced liver fibrosis in rats. Br J Pharmacol 2016;173:3248-3260.
    Pubmed KoreaMed CrossRef
  47. Avila DV, Barker DF, Zhang J, McClain CJ, Barve S, Gobejishvili L. Dysregulation of hepatic cAMP levels via altered Pde4b expression plays a critical role in alcohol-induced steatosis. J Pathol 2016;240:96-107.
    Pubmed KoreaMed CrossRef
  48. Toda K, Kumagai N, Kaneko F, et al. Pentoxifylline prevents pig serum-induced rat liver fibrosis by inhibiting interleukin-6 production. J Gastroenterol Hepatol 2009;24:860-865.
    Pubmed CrossRef
  49. DiRuggiero M, Mancuso-Stewart E, DiRuggiero D, Zirwas M. New non-steroidal topical therapies for inflammatory dermatoses: part 3: roflumilast. Skinmed 2023;21:264-268.
  50. Gupta AK, Wang T, Polla Ravi S, Bamimore MA, Piguet V, Tosti A. Systematic review of newer agents for the management of alopecia areata in adults: Janus kinase inhibitors, biologics and phosphodiesterase-4 inhibitors. J Eur Acad Dermatol Venereol 2023;37:666-679.
    Pubmed CrossRef
  51. Li MJ, Briones MS, Heinzerling KG, Kalmin MM, Shoptaw SJ. Ibudilast attenuates peripheral inflammatory effects of methamphetamine in patients with methamphetamine use disorder. Drug Alcohol Depend 2020;206:107776.
    Pubmed KoreaMed CrossRef
  52. Sid B, Verrax J, Calderon PB. Role of AMPK activation in oxidative cell damage: implications for alcohol-induced liver disease. Biochem Pharmacol 2013;86:200-209.
    Pubmed CrossRef
  53. Day EA, Ford RJ, Steinberg GR. AMPK as a therapeutic target for treating metabolic diseases. Trends Endocrinol Metab 2017;28:545-560.
    Pubmed CrossRef
  54. You M, Matsumoto M, Pacold CM, Cho WK, Crabb DW. The role of AMP-activated protein kinase in the action of ethanol in the liver. Gastroenterology 2004;127:1798-1808.
    Pubmed CrossRef
  55. Choi BK, Kim TW, Lee DR, et al. A polymethoxy flavonoids-rich Citrus aurantium extract ameliorates ethanol-induced liver injury through modulation of AMPK and Nrf2-related signals in a binge drinking mouse model. Phytother Res 2015;29:1577-1584.
    Pubmed CrossRef
  56. Wang PY, Feng JY, Zhang Z, et al. The adipokine orosomucoid alleviates adipose tissue fibrosis via the AMPK pathway. Acta Pharmacol Sin 2022;43:367-375.
    Pubmed KoreaMed CrossRef
  57. El Awdan SA, Amin MM, Hassan A. Cilostazol attenuates indices of liver damage induced by thioacetamide in albino rats through regulating inflammatory cytokines and apoptotic biomarkers. Eur J Pharmacol 2018;822:168-176.
    Pubmed CrossRef
  58. Abdel Kawy HS. Cilostazol attenuates cholestatic liver injury and its complications in common bile duct ligated rats. Eur J Pharmacol 2015;752:8-17.
    Pubmed CrossRef
  59. Han K, Zhang Y, Yang Z. Cilostazol protects rats against alcohol-induced hepatic fibrosis via suppression of TGF-β1/CTGF activation and the cAMP/Epac1 pathway. Exp Ther Med 2019;17:2381-2388.
    Pubmed KoreaMed CrossRef
  60. Cahill A, Cunningham CC, Adachi M, et al. Effects of alcohol and oxidative stress on liver pathology: the role of the mitochondrion. Alcohol Clin Exp Res 2002;26:907-915.
    Pubmed KoreaMed CrossRef
  61. Insel PA, Murray F, Yokoyama U, et al. cAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol 2012;166:447-456.
    Pubmed KoreaMed CrossRef
  62. Shimizu E, Kobayashi Y, Oki Y, Kawasaki T, Yoshimi T, Nakamura H. OPC-13013, a cyclic nucleotide phosphodiesterase type III, inhibitor, inhibits cell proliferation and transdifferentiation of cultured rat hepatic stellate cells. Life Sci 1999;64:2081-2088.
    Pubmed CrossRef
  63. El Awdan SA, Abdel Rahman RF, Ibrahim HM, et al. Regression of fibrosis by cilostazol in a rat model of thioacetamide-induced liver fibrosis: up regulation of hepatic cAMP, and modulation of inflammatory, oxidative stress and apoptotic biomarkers. PLoS One 2019;14:e0216301.
    Pubmed KoreaMed CrossRef
  64. Saito S, Hata K, Iwaisako K, et al. Cilostazol attenuates hepatic stellate cell activation and protects mice against carbon tetrachloride-induced liver fibrosis. Hepatol Res 2014;44:460-473.
    Pubmed CrossRef
  65. Oh YJ, Kim HY, Lee MH, et al. Cilostazol improves HFD-induced hepatic steatosis by upregulating hepatic STAMP2 expression through AMPK. Mol Pharmacol 2018;94:1401-1411.
    Pubmed CrossRef
  66. Wada T, Onogi Y, Kimura Y, et al. Cilostazol ameliorates systemic insulin resistance in diabetic db/db mice by suppressing chronic inflammation in adipose tissue via modulation of both adipocyte and macrophage functions. Eur J Pharmacol 2013;707:120-129.
    Pubmed CrossRef
  67. Aoki C, Hattori Y, Tomizawa A, Jojima T, Kasai K. Anti-inflammatory role of cilostazol in vascular smooth muscle cells in vitro and in vivo. J Atheroscler Thromb 2010;17:503-509.
    Pubmed CrossRef
  68. Jeon BH, Lee YH, Yun MR, et al. Increased expression of ATP-binding cassette transporter A1 (ABCA1) as a possible mechanism for the protective effect of cilostazol against hepatic steatosis. Metabolism 2015;64:1444-1453.
    Pubmed CrossRef
  69. Chen Y, Pandiri I, Joe Y, et al. Synergistic effects of cilostazol and probucol on ER stress-induced hepatic steatosis via heme oxygenase-1-dependent activation of mitochondrial biogenesis. Oxid Med Cell Longev 2016;2016:3949813.
    Pubmed KoreaMed CrossRef
  70. Ikeda Y, Matsumata T, Takenaka K, Yamagata M, Sugimachi K. Effects of doxorubicin and/or cilostazol on cancer cells during liver regeneration after two-thirds hepatectomy in rats. Oncology 1998;55:354-356.
    Pubmed CrossRef
  71. von Heesen M, Dold S, Müller S, et al. Cilostazol improves hepatic blood perfusion, microcirculation, and liver regeneration after major hepatectomy in rats. Liver Transpl 2015;21:792-800.
    Pubmed CrossRef
  72. Sugita H, Nakanuma S, Munesue S, et al. Cilostazol improves the prognosis after hepatectomy in rats with sinusoidal obstruction syndrome. J Gastroenterol Hepatol 2024;39:1413-1421.
    Pubmed CrossRef
  73. Joe Y, Zheng M, Kim HJ, et al. Cilostazol attenuates murine hepatic ischemia and reperfusion injury via heme oxygenase-dependent activation of mitochondrial biogenesis. Am J Physiol Gastrointest Liver Physiol 2015;309:G21-G29.
    Pubmed CrossRef
  74. Fujii T, Obara H, Matsubara K, et al. Oral administration of cilostazol improves survival rate after rat liver ischemia/reperfusion injury. J Surg Res 2017;213:207-214.
    Pubmed CrossRef
  75. von Heesen M, Müller S, Keppler U, et al. Preconditioning by cilostazol protects against cold hepatic ischemia-reperfusion injury. Ann Transplant 2015;20:160-168.
    Pubmed CrossRef
Gut and Liver

Vol.19 No.1
January, 2025

pISSN 1976-2283
eISSN 2005-1212

qrcode
qrcode

Share this article on :

  • line

Popular Keywords

Gut and LiverQR code Download
qr-code

Editorial Office