Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

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

  • Sally A. El Awdan,

    Roles Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Writing – original draft

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

  • Rehab F. Abdel Rahman,

    Roles Investigation, Methodology, Writing – original draft

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

  • Heba M. Ibrahim,

    Roles Investigation, Methodology, Writing – original draft

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

  • Rehab R. Hegazy ,

    Roles Investigation, Methodology, Writing – original draft, Writing – review & editing

    rehab_hegazy@hotmail.com

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

  • Salma A. El Marasy,

    Roles Investigation, Methodology

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

  • Manal Badawi,

    Roles Investigation

    Affiliation Pathology Department, Medical Division, National Research Centre, Giza, Egypt

  • Mahmoud S. Arbid

    Roles Supervision

    Affiliation Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt

Abstract

In liver fibrosis, conversion of fibroblasts to profibrogenic myofibroblasts significantly drives the development of the disease. A crucial role of cyclic adenosine monophosphate (cAMP) in regulation of fibroblast function has been reported. Increase in cAMP levels has been found to decrease fibroblast proliferation, inhibit their conversion to myofibroblast, and stimulate their death. cAMP is generated by adenyl cyclase (AC), and degraded by cyclic nucleotide phosphodiesterase (PDE). In this study, the antifibrotic effect of a PDE inhibitor, cilostazol (Cilo), on a rat model of liver fibrosis induced by thioacetamide (TAA) was investigated. Four groups of rats were used; the first group received the vehicles and served as the normal control group, while liver fibrosis was induced in the other groups using (TAA, 200 mg/kg/biweekly for 8 successive weeks, ip). The last two groups were treated with Cilo (50 and 100 mg/kg/day, po, respectively). Induction of liver fibrosis in TAA-treated rats was observed as evidenced by the biochemical and histopathological findings. On the other hand, a potent antifibrotic effect was observed in the groups treated with Cilo, with preference to the higher dose. In these groups, a significant increase in the liver content of cAMP was demonstrated that was accompanied by reduction in the hepatic expression of key fibrogenic cytokines, growth factors, and inflammatory biomarkers, including interleukin-6, tumor necrosis factor-alpha, nuclear factor kappa B, and transforming growth factor-beta as compared to TAA group. Moreover, amelioration of TAA-induced oxidative stress and apoptosis in the liver has been observed. These findings reveal the antifibrotic effect of Cilo against TAA-induced liver fibrosis in rats, and suggest regulation of cAMP pathway, together with the modulation of oxidative stress, inflammation, and apoptosis as mechanistic cassette underlines this effect.

Introduction

Liver fibrosis is a major health problem that threatens life and results in a significant percent in morbidity and mortality [1]. It is a reversible wound-healing response that occurs as a result of acute or chronic liver injury. The pathogenesis of liver injury is complex and it is characterized by the deposition of extracellular matrix (ECM) [24]. All liver cell types has their own role in the development of liver fibrosis, and it is now believed that there is a cross talk between cells of different types in the process of fibrosis through specific mediators released as interleukins, growth factors and reactive oxygen species (ROS) [5]. Many strategies have been made to attenuate liver fibrosis by inhibiting pathways or inhibiting certain molecular targets involved in the development of liver fibrosis [6].

In contrast, some protective molecular pathways become repressed during the course of liver injury. One of these important including signaling pathways is the cyclic adenosine monophosphate (cAMP) pathway [7]. cAMP is a second messenger that plays a central role in cellular responses to neurotransmitters and hormones [8]. It regulates various cellular functions including inflammation and cell differentiation by affecting gene/protein expression and function [9]. Increasing cAMP levels has been found to inhibit the conversion of resting fibroblasts/fibroblast-like cells (such as hepatic stellate cells, HSC) to profibrogenic myofibroblasts after cell injury [10], decrease their proliferation [11], stimulate their death [12], and inhibit ECM protein synthesis [13, 14]. Hence, cAMP pathway is a potential target to blunt fibrosis [6].

cAMP is generated by adenyl cyclase (AC) in response to activation of G-protein-coupled receptors, and degraded by cyclic nucleotide phosphodiesterase (PDE) into adenosine monophosphate; thus, both AC and PDE regulate the intracellular level of cAMP [15]. Cilostazol (Cilo) is a selective type-3 PDE inhibitor that is used to inhibit platelet aggregation by increasing the intracellular cAMP. Besides, Cilo showed other cAMP pathway-dependent and -independent pharmacological effects including anti-inflammatory [16, 17], antioxidant [18], and anti-apoptotic actions [19, 20]. A beneficial effect of Cilo against some rodents models of liver injury induced by ethanol [19], galactosamine [21], ischemia reperfusion [22], and carbon tetrachloride (CCl4) [23] has been demonstrated. However, its potential hepatoprotective effect against thioacetamide (TAA)-induced hepatic fibrogenesis in rats has not been evaluated, which is the aim of the current experiment, along with the study of the possible mechanistic scenario.

Materials and methods

Drugs and chemicals

TAA was purchased from Sigma-Aldrich Co. (USA) and was dissolved in 0.9% w/v saline solution for intraperitoneal (ip) injection. Cilo was purchased from Otsuka Pharmaceuticals (UK), and used orally (po). All other chemical reagents were purchased from Sigma-Aldrich chemical Co. (USA).

Experimental animals

Animals used were adult male Wister rats, weighing 150–200 g each, purchased from the Animal House at the National Research Centre (NRC, Egypt). Rats were maintained under standard conditions of temperature (25 °C) with a 12 h (light)– 12 h (dark) cycle, and were allowed free access to standard laboratory food and water. This study has been approved by the ethics committees of the Committee of Animal Care and Use of NRC (Egypt). All procedures and experiments were performed according to the protocol approved by them, and the animals were treated according to the national and international ethics guidelines. The earliest scientifically justified endpoint was used in this study to prevent pain or distress in the experimental animals. Rats were sacrificed by cervical dislocation under ether anesthesia.

Induction of liver fibrosis and experimental design

Rats were randomly allocated into four groups (8 animals per group), and treated for eight consecutive weeks according to the following scheme:

  1. The normal control group, where rats received saline ip twice weekly, and the vehicle orally once daily.
  2. The fibrotic control (TAA group), where rats received TAA (200 mg/kg, ip) twice weekly to provoke liver fibrosis [24], and received the vehicle orally once daily.
  3. TAA-Cilo50 group, where rats received Cilo (50 mg/kg/day, po) [25] for eight consecutive weeks, together with injection of TAA twice weekly.
  4. TAA-Cilo100 group, where rats received Cilo (100 mg/kg/day, po) [25] for eight consecutive weeks, together with injection of TAA twice weekly.

Twenty-four hours following the last drug administration, blood samples were withdrawn from the retro-orbital plexus of the rats under light ether anesthesia. Then, rats were sacrificed by cervical dislocation under the same anesthesia for collection of liver samples. A weighed part of the liver of each animal was rapidly dissected out, washed and homogenized using phosphate-buffered saline (PBS, 50 mM potassium phosphate, pH 7.5) at 4°C to produce a 20% homogenate. Liver homogenates were kept at -80°C till time of analysis. Another part of liver tissues were kept in 10% formalin-saline for histopathological examination.

Assessment of liver functions biomarkers

Levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were determined colorimetrically using commercially available kits (Biodiagnostic, Egypt) [26].

Assessment of oxidative stress markers

Reduced glutathione (GSH) was determined in the liver homogenate with Ellman’s reagent according to a previously described method [27]. In addition, the products of lipid peroxidation (mainly malondialdehyde, MDA) were determined as thiobarbituric acid-reactive substances [28].

Enzyme-linked immunosorbent assay (ELISA) of some biomarkers in liver homogenate

Liver homogenates were assayed for cAMP using specific Rat ELISA kit (R&D Systems, USA; Catalogs Number: KGE012B).

Tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and nuclear factor-kappa B (NF-κB) as important signals in liver injury [29], were also assayed, as well as transforming growth factor-beta (TGF-β) as a fibrogenesis-driving cytokine [30], using specific Rat ELISA kits (SunLong Biotech Co., LTD, China; Catalogs Number: SL0722Ra, SL0411Ra, SL0537Ra, and SL0705Ra, respectively).

As caspase-3 is involved in the apoptosis of hepatocytes during liver injury [31], CUSABIO Rat Casp-3 ELISA Kit (USA; Catalog Number: CSB-E08857r) was used to measure the level of caspase-3 in liver homogenate. Moreover, evaluation of α-smooth muscle actin (α-SMA) and type I collagen (COL-1) as classic liver fibrosis markers was also performed using specific Rat ELISA kits (CUSABIO, USA; Catalogs Number: CSB-E 14407r and CSB-E10134r, respectively).

Histopathological examination

For histopathological examination, morphometry and fibrosis assessment, liver tissues were immediately fixed to 10% formal saline for paraffin embedding, sectioning, and staining with Hematoxylin & Eosin (H&E) and Masson trichrome stains. The Masson trichrome specifically stain the collagen to give blue color staining of the fibrous tissue. Quantitative analysis was done in the Pathology Department of the NRC, using the Leica Qwin 500 Image Analyzer (LEICA Imaging Systems Ltd, Cambridge, England).

Ten random fields were examined for estimation of fibrous tissue percent, portal tract area (μm2), inflammatory cell infiltration, and hepatocytic degeneration. The mean from these ten fields was calculated which represent the histopathological condition of the liver tissue.

Statistical analysis

All the values are presented as means ± standard error of the means (SE), n = 8. Comparisons between different groups were carried out using one-way analysis of variance (ANOVA) followed by Tukey-Kramer test for multiple comparisons. Graphpad Prism software, version 5 (USA) was used to carry out these statistical tests. The difference was considered significant when p ˂ 0.05.

Results

Effect of Cilo on the serum AST and ALT levels in TAA-induced liver fibrosis in rats

Injection of TAA resulted in a considerable elevation in serum ALT and AST levels as compared to normal group values (1.85 and 1.57 fold, respectively). Both Clio-treated groups had a significant decrease in serum ALT and AST when compared to TAA control group. Cilo 50 and 100 mg/kg depleted the raised ALT levels by 27%, and 35%, respectively, as compared to TAA control group. Moreover, cilostazol 50 and 100 mg/kg depleted the raised AST levels by 24%, and 21%, respectively as compared to TAA control group (Fig 1).

thumbnail
Fig 1. Effect of cilostazol on the serum ALT (A), AST (B) levels in rats with TAA-induced liver fibrosis.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); ALT, alanine aminotransferase; AST, aspartate aminotransferase. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g001

Effect of Cilo on the liver contents of oxidative stress markers in TAA-induced fibrosis in rats

Injection of TAA resulted in a significant depletion in reduced glutathione (GSH) levels (24%) (Fig 2A) as well as a significant elevation in MDA values (50%) as compared to normal group values (Fig 2B). On the other hand, Cilo 50 and 100 mg/kg raised the depleted GSH levels by 16% and 21%, respectively, while decreased the raised MDA levels by 4% and 30%, respectively, as compared to TAA control group.

thumbnail
Fig 2. Effect of cilostazol on liver GSH (A) and MDA (B) in TAA-induced fibrosis in rats.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); GSH: reduced glutathione, MDA, malondialdehyde. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g002

Effect of Cilo on the liver content of cAMP in TAA-induced fibrosis in rats

A significant reduction of the normal hepatic content of cAMP was observed in the rats with TAA-induced liver fibrosis. However, a dose-dependent increase of this content was observed in the groups treated with Cilo, 50 and 100 mg/kg (Fig 3).

thumbnail
Fig 3. Effect of cilostazol on liver content of cAMP in TAA-induced fibrosis in rats.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); cAMP: cyclic adenosine monophosphate. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g003

Effect of Cilo on the hepatic contents of some cytokines and chemokines in TAA-induced fibrosis in rats

Injection of TAA resulted in a significant elevations in liver TNF-α, IL-6, NFkβ and TGF-β (2.14, 2.1, 2.13 and 2.3 fold respectively) as compared to normal group values.

Only Cilo 100 mg/kg significantly decreased the raised TNF-α and TGF-β hepatic levels as compared to TAA control group. On the other hand, both doses of cilo, 50 and 100 mg/kg, significantly decreased the raised IL-6 levels by 33% and 31%, respectively, as well as NF-kβ levels by 27 and 56%, respectively, comparing to TAA control group (Fig 4).

thumbnail
Fig 4. Effect of cilostazol on liver TNF-α (A), IL-6 (B), NFκβ (C) and TGFβ (D) in TAA-induced fibrosis in rats.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); TNF-α: tumor necrosis factor α; IL-6, interleukin-6; NF-κB, nuclear factor-kappa B; TGF-β, transforming growth factor-beta. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g004

Effect of Cilo on caspase-3 in TAA-induced fibrosis in rats

Induction of liver fibrosis in rats using TAA resulted in a significant elevation in hepatic caspase-3 (4.7-fold) as compared to normal group value (Fig 5). Treatment of rats with Cilo, 50 and 100 mg/kg, significantly decreased the raised hepatic caspase-3 levels by 44% and 62%, respectively, as compared to TAA group.

thumbnail
Fig 5. Effect of cilostazol on liver caspase-3 in TAA-induced fibrosis in rats.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); Caspase 3. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g005

Effect of Cilo on α-SMA and COL-1 in TAA-induced fibrosis in rats

Induction of liver fibrosis in rats using TAA resulted in a significant elevation in hepatic α-SMA and COL-1 as compared to the normal group (Fig 6). Treatment of rats with Cilo, 50 and 100 mg/kg, significantly decreased the raised hepatic α-SMA and COL-1 levels comparing with TAA group.

thumbnail
Fig 6. Effect of cilostazol on liver α-SMA (A) and COL-1 (B) in TAA-induced fibrosis in rats.

Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po); α-SMA, α-smooth muscle actin; COL-1, type I collagen. Data presented as mean ± S.E.; n = 8. * Significantly different from Normal group at p˂0.05. @ Significantly different from TAA group at p˂0.05.

https://doi.org/10.1371/journal.pone.0216301.g006

Effect of Cilo on the liver histopathological findings in TAA-induced fibrosis in rats

Liver of normal rats revealed normal hepatic parenchyma with normal hepatocytes (Fig 7a) and scant faint blue-stained fibrous tissue demonstrated in the portal area (Fig 8a). On the contrary, disrupted hepatic parenchyma, with massive fibrous tissue proliferation extending from the portal area, associated with ballooning degeneration of hepatocytes in addition to apoptosis, was demonstrated in TAA-treated group (Fig 7b). Atypical biliary epithelial hyperplasia associated with leukocytic cell infiltration was characteristically demonstrated in this group. The collagen fibers were strongly blue in Masson´s trichrome stained sections (Fig 8b). Diminution of fibrosis with minimal inflammatory infiltrates were demonstrated in TAA-Cilo50-treated group (Figs 7c and 8c). Interestingly, pronounced amelioration was demonstrated in TAA-Cilo100-treated group, in which the hepatic parenchyma in most examined sections appeared normal with scarce fibrous tissue confined to the portal area (Figs 7d and 8d) (Table 1).

thumbnail
Fig 7. liver tissue, stained with H&E, of (a) normal rats showing normal hepatic parenchyma with normal hepatocytes, (b) TAA-treated group showing disrupted hepatic parenchyma, with massive fibrous tissue proliferation extending from the portal area, associated with ballooning degeneration of hepatocytes in addition to apoptosis, (c) TAA-Cilo50-treated group showing diminished fibrosis with minimal inflammatory infiltrates, and (d) TAA-Cilo100-treated group showing normal preserved parenchyma.

(H&E, 20X). Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po).

https://doi.org/10.1371/journal.pone.0216301.g007

thumbnail
Fig 8. liver tissue, stained with Masson´s trichrome staine, of (a) normal rats showing scant faint blue-stained fibrous in the portal area, (b) TAA-treated group showing strong blue-stained collagen fiber, (c) TAA-Cilo50-treated group showing diminished blue-stained collagen fibers, and (d) TAA-Cilo100-treated group showing scarce blue-stained fibrous tissue confined to the portal area.

(Masson´s trichrome staine, 20X). Normal, rats treated with vehicles; TAA, rats treated with thioacetamide (200 mg/kg/biweekly for 8 weeks, ip); TAA-Clio, rats treated with thioacetamide and cilostazol (50 or 100 mg/kg/day for 8 weeks, po).

https://doi.org/10.1371/journal.pone.0216301.g008

thumbnail
Table 1. Histopathological parameters of the liver in the studied groups.

https://doi.org/10.1371/journal.pone.0216301.t001

Discussion

The current study has targeted evaluation of Cilo as a therapeutic alternative to liver fibrosis and analyzing the mechanism by which it acts to attenuate liver fibrosis elicited in rats by TAA. Induction of liver fibrosis by TAA is occurred as a result of its biotransformation, by CYP2E1 enzymes in the liver cells’ microsomes, to a very reactive intermediate known as TAA sulphur dioxide through oxidation [32]. This results in variable grades of hepatotoxicity including nodular cirrhosis, proliferation of hepatic cells, and necrosis of parenchyma cells [33]. TA is a common hepatotoxicant model to study liver injury due to its outstanding solubility in the water, and a prolonged injury and recovery pattern giving significant time to study mechanisms [34, 35]. Unlike CCl4, TA toxicity does not depend on induction of oxidative stress only [36]. Therefore, using TAA-induced liver fibrosis model allows assessing different pathophysiological factors, parameters and mechanisms that may underline the progression of liver diseases rather than oxidative stress only. This adds an advantage to the TAA-hepatotoxic model by its potential to simulate some clinical conditions where many precipitating factors are involved in the development of liver fibrosis, and different mechanisms underline its progression [24]. Moreover, because of its limited use as a model toxicant comparing to other experimental animal models, especially in the chronic studies, the exact mechanism of TA-induced necrotic cell death is not clear. Thus, using it in more studies investigating different pathways are required and would add to the knowledge.

In the present study, TAA-induced liver damage in rats was evidenced, biochemically, by the alterations in serum levels of ALT and AST, as well as hepatic fibrosis biomarkers in TAA-induced group, and confirmed by the histopathological findings. Moreover, induction of oxidative stress state in the current TAA-treated group has been proved by the decline in the normal liver GSH content as well as an elevation in MDA content. These results are in accordance with previous studies showed that TAA, by its oxidative stress capacity that exceeds the capacity of body’s antioxidative and protective mechanisms [37], damages liver cell membranes, resulting in the leakage of the cytoplasmic liver enzymes ALT and AST into blood stream in amounts related to the extent of liver damage [3840].

Administration of Cilo in the current study significantly improved the liver function, oxidative stress, and liver fibrosis biomarkers, together with the histopathological findings, when compared to the control TAA group; this was in preference to the higher dose (100 mg/kg). These findings indicate the beneficial effects of Cilo in the healing and regeneration of the hepatocytes, and show its antioxidant potential [18]. Same effects of Cilo have been shown before when other research groups used other rodent’s models of liver fibrosis induced by ethanol and CCl4 [19, 23]. Other PDE inhibitor, pentoxifylline, has shown similar results against TAA-induced model [41].

In the current TAA-induced liver fibrosis model, reduction of the hepatic cAMP content was observed. This support the data that cAMP pathway is considered as protective signaling pathway against liver fibrosis that become repressed during liver injury [7]. In line with that, the increased level of cAMP observed in the liver of the rats treated with the PDE inhibitor, Cilo, justifies its hepatoprotective effects.

The liver is considered as a main organ controlling cytokines activity and its production depending on the initial early pro-inflammatory cytokines released from macrophages [42, 43]. TAA, as a hepatotoxin, induces pro-inflammatory and inflammatory cytokines and mediators by macrophages (Kupffer cells) such as TNF-α, IL-6, NF-κβ and TGFβ that play important roles in hepatic inflammation [44, 45]. The findings of our study are in line with these previous observations. NF-κB plays an important role in the expression of many pro-inflammatory genes [46], and its induction has been found to play a crucial role in TAA-induced liver fibrosis [47, 48]. After activation with pro-inflammatory cytokines, NF-ĸB acts as the driving force of fibrosis. It activates TGFβ, which is considered as the key mediator in fibrogenesis as it potentiates the conversion of HSCs into myofibroblasts that stimulates the synthesis of ECM proteins, and hinders its degradation [49]. An increase in the relative genetic expression of TGFβ due to TAA administration has been reported [50, 51].

Many studies have shown that HSCs activation can be attenuated by blocking the NFκβ signaling pathway [52, 53]. In our study, Cilo, in a dose-dependent manner, significantly reduced the hepatic proinflammatory cytokines, TNF-α and IL-6, contents, as well as NFκβ and TGFβ1 levels when compared to TAA-induced group. These findings suggest that one of the possible mechanisms involved in the antifibrotic effect of Cilo might be through its previously reported anti-inflammatory action [16, 17] with suppression of proinflamatory and fibrogenic cytokines.

Moreover, elevation of caspase-3, as a marker of apoptosis, was odserved in the liver of TAA-treated rats. Fourteen caspases have been involved in the apoptotic pathway cascade [54]. Among these, caspase-3 is considered to be a major execution protease. Caspase-3 is a sensitive marker reflecting liver damage and is associated with liver fibrosis as well [55]. It is suggested that the inflammation that occurred in the liver cells elicited by TAA lead in the later stages to apoptosis of hepatocytes reflected by the elevated level of caspase-3 in the TAA-induced rats. Cilo in both doses significantly depleted caspase-3 level elevation, indication a protective and anti-apoptotic effect of Cilo. This observation is in agreement with other data reported the anti-apoptotic effect of Cilo [56, 57].

Conclusion

These findings revealed that the PDE inhibitor Cilo showed hepatoprotective effects against TAA-induced liver fibrosis in rats. It modulated many process in the liver, including the oxidative stress, cAMP pathway, inflammation, ECM and collagen deposition, as well as apoptosis. The study proposes that these effects worked together to reduce fibrogenesis, and suggest Cilo as a promising antifibrotic agent against liver fibrosis. More comprehensive mechanistic studies are recommended to bring to light the antifibrotic role of Cilo in liver injury.

Supporting information

S1 File. Raw data-PLOS one-Elawdan et al.docx.

https://doi.org/10.1371/journal.pone.0216301.s001

(DOCX)

Acknowledgments

SE, as a principle investigator, was funded by the National Research Centre (Egypt, http://www.nrc.sci.eg/) to perform this study, Grant no. 11010127. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  1. 1. Sanchez-Valle V, Chavez-Tapia NC, Uribe M, Mendez-Sanchez N. Role of oxidative stress and molecular changes in liver fibrosis: a review. Current medicinal chemistry`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  2. 2. Greenwel P, Dominguez-Rosales JA, Mavi G, Rivas-Estilla AM, Rojkind M. Hydrogen peroxide: a link between acetaldehyde-elicited alpha1(I) collagen gene up-regulation and oxidative stress in mouse hepatic stellate cells. Hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  3. 3. Pinzani M, Macias-Barragan J. Update on the pathophysiology of liver fibrosis. Expert review of gastroenterology & hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  4. 4. Alegre F, Pelegrin P, Feldstein AE. Inflammasomes in Liver Fibrosis. Seminars in liver disease`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  5. 5. Kmiec Z. Cooperation of liver cells in health and disease. Advances in anatomy, embryology, and cell biology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  6. 6. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nature reviews Molecular cell biology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  7. 7. Wahlang B, McClain C, Barve S, Gobejishvili L. Role of cAMP and phosphodiesterase signaling in liver health and disease. Cellular signalling`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  8. 8. Sutherland EW, Rall TW. Fractionation and characterization of a cyclic adenine ribonucleotide formed by tissue particles. The Journal of biological chemistry`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  9. 9. Dal Piaz V, Giovannoni MP. Phosphodiesterase 4 inhibitors, structurally unrelated to rolipram, as promising agents for the treatment of asthma and other pathologies. European journal of medicinal chemistry`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  10. 10. Swaney JS, Roth DM, Olson ER, Naugle JE, Meszaros JG, Insel PA. Inhibition of cardiac myofibroblast formation and collagen synthesis by activation and overexpression of adenylyl cyclase. Proceedings of the National Academy of Sciences of the United States of America`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  11. 11. Stumm CL, Wettlaufer SH, Jancar S, Peters-Golden M. Airway remodeling in murine asthma correlates with a defect in PGE2 synthesis by lung fibroblasts. American journal of physiology Lung cellular and molecular physiology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  12. 12. Zhang P, Su J, King ME, Maldonado AE, Park C, Mende U. Regulator of G protein signaling 2 is a functionally important negative regulator of angiotensin II-induced cardiac fibroblast responses. American journal of physiology Heart and circulatory physiology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  13. 13. Insel PA, Murray F, Yokoyama U, Romano S, Yun H, Brown L, et al. cAMP and Epac in the regulation of tissue fibrosis. British journal of pharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  14. 14. Miller CL, Cai Y, Oikawa M, Thomas T, Dostmann WR, Zaccolo M, et al. Cyclic nucleotide phosphodiesterase 1A: a key regulator of cardiac fibroblast activation and extracellular matrix remodeling in the heart. Basic research in cardiology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  15. 15. McKnight GS. Cyclic AMP second messenger systems. Current opinion in cell biology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  16. 16. da Motta NA, de Brito FC. Cilostazol exerts antiplatelet and anti-inflammatory effects through AMPK activation and NF-kB inhibition on hypercholesterolemic rats. Fundamental & clinical pharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  17. 17. Sakamoto T, Ohashi W, Tomita K, Hattori K, Matsuda N, Hattori Y. Anti-inflammatory properties of cilostazol: Its interruption of DNA binding activity of NF-kappaB from the Toll-like receptor signaling pathways. International immunopharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  18. 18. Kurtoglu T, Basoglu H, Ozkisacik EA, Cetin NK, Tataroglu C, Yenisey C, et al. Effects of cilostazol on oxidative stress, systemic cytokine release, and spinal cord injury in a rat model of transient aortic occlusion. Annals of vascular surgery`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  19. 19. Lee YJ, Eun JR. Cilostazol Decreases Ethanol-Mediated TNFalpha Expression in RAW264.7 Murine Macrophage and in Liver from Binge Drinking Mice. The Korean journal of physiology & pharmacology: official journal of the Korean Physiological Society and the Korean Society of Pharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  20. 20. Choi JM, Shin HK, Kim KY, Lee JH, Hong KW. Neuroprotective effect of cilostazol against focal cerebral ischemia via antiapoptotic action in rats. The Journal of pharmacology and experimental therapeutics`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  21. 21. Taye A, El-Moselhy MA, Hassan MK, Ibrahim HM, Mohammed AF. Hepatoprotective effect of pentoxifylline against D-galactosamine-induced hepatotoxicity in rats. Annals of hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  22. 22. Amasyali AS, Akkurt A, Kazan E, Yilmaz M, Erol B, Yildiz Y, et al. The protective effect of tadalafil on IMA (ischemia modified albumin) levels in experimental renal ischemia-reperfusion injury. International journal of clinical and experimental medicine`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  23. 23. Saito S, Hata K, Iwaisako K, Yanagida A, Takeiri M, Tanaka H, et al. Cilostazol attenuates hepatic stellate cell activation and protects mice against carbon tetrachloride-induced liver fibrosis. Hepatology research: the official journal of the Japan Society of Hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  24. 24. Nakajima M, Iwata K, Yamamoto T, Funae Y, Yoshida T, Kuroiwa Y. Nicotine metabolism in liver microsomes from rats with acute hepatitis or cirrhosis. Drug metabolism and disposition: the biological fate of chemicals`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  25. 25. Ragab D, Abdallah DM, El-Abhar HS. Cilostazol renoprotective effect: modulation of PPAR-gamma, NGAL, KIM-1 and IL-18 underlies its novel effect in a model of ischemia-reperfusion. PloS one`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  26. 26. Reitman S, Frankel S. A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. American journal of clinical pathology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  27. 27. Beutler E, Duron O, Kelly BM. Improved method for the determination of blood glutathione. The Journal of laboratory and clinical medicine`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  28. 28. Mihara M, Uchiyama M. Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Analytical biochemistry`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  29. 29. Mandrekar P, Szabo G. Signalling pathways in alcohol-induced liver inflammation. Journal of hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  30. 30. Gressner OA, Weiskirchen R, Gressner AM. Evolving concepts of liver fibrogenesis provide new diagnostic and therapeutic options. Comparative hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  31. 31. Elsharkawy AM, Oakley F, Mann DA. The role and regulation of hepatic stellate cell apoptosis in reversal of liver fibrosis. Apoptosis: an international journal on programmed cell death`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  32. 32. Kim KH, Bae JH, Cha SW, Han SS, Park KH, Jeong TC. Role of metabolic activation by cytochrome P450 in thioacetamide-induced suppression of antibody response in male BALB/c mice. Toxicology letters`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  33. 33. Sadasivan S, Latha PG, Sasikumar JM, Rajashekaran S, Shyamal S, Shine VJ. Hepatoprotective studies on Hedyotis corymbosa (L.) Lam. Journal of ethnopharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  34. 34. Apte UM, Limaye PB, Ramaiah SK, Vaidya VS, Bucci TJ, Warbritton A, et al. Upregulated promitogenic signaling via cytokines and growth factors: potential mechanism of robust liver tissue repair in calorie-restricted rats upon toxic challenge. Toxicological sciences: an official journal of the Society of Toxicology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  35. 35. Yanguas SC, Cogliati B, Willebrords J, Maes M, Colle I, van den Bossche B, et al. Experimental models of liver fibrosis. Archives of toxicology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  36. 36. Chilakapati J, Shankar K, Korrapati MC, Hill RA, Mehendale HM. Saturation toxicokinetics of thioacetamide: role in initiation of liver injury. Drug metabolism and disposition: the biological fate of chemicals`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  37. 37. Pallottini V, Martini C, Bassi AM, Romano P, Nanni G, Trentalance A. Rat HMGCoA reductase activation in thioacetamide-induced liver injury is related to an increased reactive oxygen species content. Journal of hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  38. 38. Ayatollahi M, Hesami Z, Jamshidzadeh A, Gramizadeh B. Antioxidant Effects of Bone Marrow Mesenchymal Stem Cell against Carbon Tetrachloride-Induced Oxidative Damage in Rat Livers. International journal of organ transplantation medicine`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  39. 39. Mustafa HN, El Awdan SA, Hegazy GA. Protective role of antioxidants on thioacetamide-induced acute hepatic encephalopathy: biochemical and ultrastructural study. Tissue & cell`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  40. 40. Nissar AU, Farrukh MR, Kaiser PJ, Rafiq RA, Afnan Q, Bhushan S, et al. Effect of N-acetyl cysteine (NAC), an organosulfur compound from Allium plants, on experimentally induced hepatic prefibrogenic events in Wistar rat. Phytomedicine: international journal of phytotherapy and phytopharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  41. 41. Luo M, Dong L, Li J, Wang Y, Shang B. Protective effects of pentoxifylline on acute liver injury induced by thioacetamide in rats. International journal of clinical and experimental pathology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  42. 42. Muriel P, Rivera-Espinoza Y. Beneficial drugs for liver diseases. Journal of applied toxicology: JAT`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  43. 43. Simpson KJ, Lukacs NW, Colletti L, Strieter RM, Kunkel SL. Cytokines and the liver. Journal of hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  44. 44. Wu YL, Lian LH, Wan Y, Nan JX. Baicalein inhibits nuclear factor-kappaB and apoptosis via c-FLIP and MAPK in D-GalN/LPS induced acute liver failure in murine models. Chemico-biological interactions`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  45. 45. Diao Y, Zhao XF, Lin JS, Wang QZ, Xu RA. Protection of the liver against CCl4-induced injury by intramuscular electrotransfer of a kallistatin-encoding plasmid. World journal of gastroenterology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  46. 46. Reyes-Gordillo K, Segovia J, Shibayama M, Vergara P, Moreno MG, Muriel P. Curcumin protects against acute liver damage in the rat by inhibiting NF-kappaB, proinflammatory cytokines production and oxidative stress. Biochimica et biophysica acta`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  47. 47. Demirel U, Yalniz M, Aygun C, Orhan C, Tuzcu M, Sahin K, et al. Allopurinol ameliorates thioacetamide-induced acute liver failure by regulating cellular redox-sensitive transcription factors in rats. Inflammation`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  48. 48. Hessin A, Hegazy R, Hassan A, Yassin N, Kenawy S. Lactoferrin Enhanced Apoptosis and Protected Against Thioacetamide-Induced Liver Fibrosis in Rats. Open access Macedonian journal of medical sciences`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  49. 49. Arendt E, Ueberham U, Bittner R, Gebhardt R, Ueberham E. Enhanced matrix degradation after withdrawal of TGF-beta1 triggers hepatocytes from apoptosis to proliferation and regeneration. Cell proliferation`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  50. 50. Guerra RR, Trotta MR, Parra OM, Avanzo JL, Bateman A, Aloia TP, et al. Modulation of extracellular matrix by nutritional hepatotrophic factors in thioacetamide-induced liver cirrhosis in the rat. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas / Sociedade Brasileira de Biofisica [et al]`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  51. 51. Foo NP, Lin SH, Lee YH, Wu MJ, Wang YJ. alpha-Lipoic acid inhibits liver fibrosis through the attenuation of ROS-triggered signaling in hepatic stellate cells activated by PDGF and TGF-beta. Toxicology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  52. 52. Ding H, Shi J, Wang Y, Guo J, Zhao J, Dong L. Neferine inhibits cultured hepatic stellate cell activation and facilitates apoptosis: A possible molecular mechanism. European journal of pharmacology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  53. 53. Chen G, Wang Y, Li M, Xu T, Wang X, Hong B, et al. Curcumol induces HSC-T6 cell death through suppression of Bcl-2: involvement of PI3K and NF-kappaB pathways. European journal of pharmaceutical sciences: official journal of the European Federation for Pharmaceutical Sciences`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  54. 54. Nicholson DW. Caspase structure, proteolytic substrates, and function during apoptotic cell death. Cell death and differentiation`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  55. 55. Bantel H, Lugering A, Heidemann J, Volkmann X, Poremba C, Strassburg CP, et al. Detection of apoptotic caspase activation in sera from patients with chronic HCV infection is associated with fibrotic liver injury. Hepatology`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  56. 56. Kim MJ, Lee JH, Park SY, Hong KW, Kim CD, Kim KY, et al. Protection from apoptotic cell death by cilostazol, phosphodiesterase type III inhibitor, via cAMP-dependent protein kinase activation. Pharmacological research`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.
  57. 57. Ba XH, Cai LP, Han W. Effect of cilostazol pretreatment on the PARP/AIF-mediated apoptotic pathway in rat cerebral ischemia-reperfusion models. Experimental and therapeutic medicine`Abbreviation. Year;|Volume|(Issue)|:Pages|. `Epub Epub Date. Accession Number.