Vous êtes sur la page 1sur 12

Pharmaceutical Biology

ISSN: 1388-0209 (Print) 1744-5116 (Online) Journal homepage: http://www.tandfonline.com/loi/iphb20

Hepatoprotective properties of extensively studied


medicinal plant active constituents: Possible
common mechanisms

Hassan Farghali, Nikolina Kutinov Canov & Samir Zakhari

To cite this article: Hassan Farghali, Nikolina Kutinov Canov & Samir Zakhari
(2015) Hepatoprotective properties of extensively studied medicinal plant active
constituents: Possible common mechanisms, Pharmaceutical Biology, 53:6, 781-791, DOI:
10.3109/13880209.2014.950387

To link to this article: http://dx.doi.org/10.3109/13880209.2014.950387

Published online: 09 Dec 2014.

Submit your article to this journal

Article views: 406

View related articles

View Crossmark data

Citing articles: 2 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iphb20

Download by: [202.67.40.194] Date: 06 October 2017, At: 09:32


http://informahealthcare.com/phb
ISSN 1388-0209 print/ISSN 1744-5116 online
Editor-in-Chief: John M. Pezzuto
Pharm Biol, 2015; 53(6): 781791
! 2014 Informa Healthcare USA, Inc. DOI: 10.3109/13880209.2014.950387

REVIEW ARTICLE

Hepatoprotective properties of extensively studied medicinal plant


active constituents: Possible common mechanisms
Hassan Farghali1, Nikolina Kutinova Canova1, and Samir Zakhari2
1
First Faculty of Medicine, Institute of Pharmacology, Charles University in Prague, Czech Republic and 2Science DISCUS, Washington, DC, USA

Abstract Keywords
Context: We focused on certain plant active constituents considered to be the most promising/ Clinical trials, evidence-based
studied for liver disease and that were critically investigated from the basic science point of hepatoprotectants, hepatotoxicity models,
view and, to some extent, the clinical one. Due to insufficient pharmacological data, most of the herbal medications, liver disease, oxidative
herbal formulations containing these molecules cannot be recommended for the treatment of stress
Downloaded by [202.67.40.194] at 09:32 06 October 2017

liver disease.
Objective: To present the most promising compounds tested experimentally and/or clinically History
and describe in brief popular models in experimental testing of potential hepatoprotective
compounds. Received 4 April 2014
Methods: A literature search using Web of Science (WOS), PubMed, and Google search was Revised 26 June 2014
performed. Accepted 25 July 2014
Results: Focusing on a few herbal hepatoprotective active constituents is useful to health Published online 9 December 2014
professionals working in the field of therapeutics to develop evidence-based hepatoprotective
agents by conducting research on pure chemical structures or on molecular modifications
using computational chemistry. This review demonstrates that multi-pathways in the liver
pathobiology can be interrupted at one or more levels by natural hepatoprotective studied,
such as interference with the oxidative stress at multiple levels to reduce reactive oxygen/
nitrogen species, resulting in ameliorating hepatotoxicity.
Conclusion: Hepatoprotective constituents of herbal medications are poorly absorbed after oral
administration; methods that can improve their bioavailability are being developed. It is
recommended that controlled prospective double-blind multicenter studies on isolated active
plant constituents, or on related newly designed molecules after structural modifications,
should be performed. This effort will lead to expanding the existing, limited drugs for the vast
majority of liver diseases.

Introduction (Rayfield, 2013). Moreover, liver diseases such as fibrosis


are a major worldwide health problem, with high prevalence
Globally, there are numerous causes for liver disease,
in developing countries where hundreds of millions are
including viral hepatitis and HIV, obesity with the consequent
afflicted (Sanchez-Valle et al., 2012). In fact, with the global
non-alcoholic fatty liver disease (NAFLD), excessive chronic
obesity epidemic, NAFLD, and the ensuing non-alcoholic
alcohol consumption, immune and cholestatic disorders,
steatohepatitis (NASH), fibrosis, cirrhosis and hepatocellular
inherited metabolic disorders, numerous medications, hemo-
carcinoma have become a worldwide health concern of all
chromatosis, schistosomiasis, and fungi infections, among
ages and ethnicities (Alonso et al., 2010; Nobili et al., 2011;
others. Liver disease is a considerable health burden across
Torres et al., 2012).
Europe (Blachier et al., 2013) and globally. In England, 2% of
In addition to regulating energy homeostasis, the liver
total deaths between 2001 and 2009 were due to liver disease
performs essential functions that sustain life, including food
(National End of Life Care Intelligence Network, 2013). The
digestion (absorption of fat and fat-soluble vitamins through
report states that, in the same period, alcoholic liver disease
bile production), metabolism, and detoxification of xeno-
accounted for well over a third (37%) of liver disease deaths.
biotics that enters the body through the digestive system or
In the United States, over 25 million Americans are afflicted
lungs, or absorbed through the skin. The liver also creates
with liver disease and more than 27 000 die from cirrhosis, the
new molecules (proteins, cytokines, etc.) that are essential for
seventh leading cause of death. Additionally, 43 000 die of
physiological and immunological functions. The liver also
liver disease, of which about 50% are alcohol related
acts as a blood filter, by converting ammonia into urea, and
by removing and excreting substances from the blood that
Correspondence: Hassan Farghali, Professor of Pharmacology, First otherwise would be toxic. In addition, the liver makes proteins
Faculty of Medicine, Institute of Pharmacology, Charles University in
Prague, Albertov 4, 128 00 Prague 2, Czech Republic. Tel/Fax: +420 that regulate blood clotting and influence the immune
224968106. E-mail: hfarg@lf1.cuni.cz system, among others. The liver has the extraordinary ability
782 H. Farghali et al. Pharm Biol, 2015; 53(6): 781791

to regenerate its own damaged tissue. This allows transplant- Rana et al., 2011; Subramanian et al., 2012; Torres-Gonalez
ing part of ones liver to a blood-related relative who is a et al., 2011; Upadhyay et al., 2013; Xavier et al., 2012; Zhang
matching type, and both people will have healthy livers that et al., 2012).
will grow to its original size. Of course, liver regeneration It is understandable that the use of substances isolated
works in healthy livers. Thus, it is imperative to highlight few from natural sources for the phytotherapy of various diseases,
aspects of liver diseases. including liver diseases, is desirable because they are
relatively inexpensive and are widely available. Silymarin
An outline of liver diseases and resveratrol are two of many examples of natural
Numerous diseases and environmental factors can affect liver substances that have shown a strong hepatoprotective poten-
function resulting in gallstones, liver steatosis, fibrosis, tial due to their antioxidant, anti-inflammatory, and liver
cirrhosis, and even cancer. By clearing xenobiotics through regenerative capabilities (Farghali et al., 2000a, 2009; Glauert
biotransformation (in most instances by detoxification or et al., 2010; Haddad et al., 2011; Pradhan & Girish, 2006).
sometimes by production of active or even more toxic Substances such as curcumin and quercetin exhibit anti-
metabolites), the liver itself can be injured by these oxidant and cytoprotective properties, but their use as
xenobiotics. Due to its high blood perfusion rate and high hepatoprotectants has not been extensively investigated
metabolic capacity, the liver is continuously exposed to high clinically. However, curcumin and quercetin exhibited sig-
levels of xenobiotics and to their metabolites. Fortunately, the nificant hepetoprotection in experimental setups as reported
liver has a high capacity of regeneration and ability to repair previously (Cerny et al., 2011; Chirumbolo, 2010; Lekic
any underlying damage. Generally speaking, hepatotoxicity et al., 2013; Zhao et al., 2011; Zhou et al., 2011). In addition,
occurs when liver regeneration capabilities are exhausted medicinal plants play a key role in human health care. The
Downloaded by [202.67.40.194] at 09:32 06 October 2017

and cell damage ensues. Hepatic injury due to xenobiotic World Health Organization (WHO) estimates that 80% of the
exposure involves inflammation, oxidative stress, and lipid populations of some Asian and African countries presently
peroxidation reactions that can result in mitochondrial use herbal medications for some aspect of primary health
damage and cell death. The resulting pathological alterations care. Studies in the United States and Europe have shown that
of the endogenous substances such as proteins, nucleic acids, the use of herbal therapy is less common in clinical settings,
and lipids can harm the proper functioning of the liver (Hong but has become increasingly more so in recent years as
et al., 2009). scientific evidence about their effectiveness has become more
widely available. Scientific studies available on medicinal
plants indicate that promising phytochemicals can be
Scientific rationale for the traditional use of some
developed for many diseases (Gupta, 1994).
plants in the management of liver diseases
In a report by Adewusi and Afolayan (2010), who
Generally, the medicinal use of plants in their natural and reviewed natural products with hepatoprotective activity,
unprocessed form began when it was first observed that more than one hundred plants with 58 compounds classified
certain food plants altered particular body functions. into appropriate chemical group were addressed. Therefore,
Historically, plants were sources of medications for millennia. these plants are attractive targets for future studies, and the
Purified active constituents such as alkaloids, glycosides, and identification of their active constituents will probably lead
flavonoids were isolated and their pharmacologic activities to new therapies for liver disease. The efficacy of herbal
were assessed, and they constituted the base for modern medications is influenced by factors such as variations in the
drug development. The Indians, Egyptians, Chinese, and soil and climate, period and place of collection, age and part
Sumerians are just a few civilizations that have provided of plants used, which influence the hepatoprotective proper-
evidence suggesting that plants can be effectively used as ties (Valan et al., 2010).
medications to treat or prevent disease. Ayurveda, a 5000- It is important to realize that despite the great advances in
year-old system of natural healing that has its origins in modern medicine, there is no effective drug available that
the Vedic culture of India described the use of food for stimulates liver function, offer liver protection, or help to
therapeutic purpose. regenerate hepatic cells (Chattopadhyay, 2003). In fact, we
Therefore, the potential use of these plants for hepatopro- need to seek novel potential hepatoprotective substances and
tection makes them an attractive target for future studies, and to search for alternative drugs for the treatment of liver
for the identification of their active constituents. This effort diseases to replace currently used medications of doubtful
may probably lead to widening the horizon of existing efficacy and safety. Notwithstanding their potential health
therapies for liver diseases. Future studies will be necessary to effects, there could be potential harm from herbal prepar-
expand the existing, limited therapeutic drugs for the vast ations either per se (Khan et al., 2008) or due to their
majority of liver diseases. Reports of medicinal plants for the interaction with conventional medicines. For instance, Bilgi
treatment of liver disease are numerous; however, due to et al. (2010) reported on a case of imatinib-induced
space limitations, we will focus on only a few (Ansari et al., exaggerated hepatotoxicity after concurrent ginseng ingestion
2011; Asgarpanah & Kazemivash, 2012; Asuku et al., 2012; in a patient with chronic myelogenous leukemia. Our interest
Avila et al., 2011; Ghosh et al., 2011; Gutierrez et al., 2013; in hepatoprotective agents from various origins whether
Haddad et al., 2011; Ho et al., 2012; Lee et al., synthetic, natural, or drugs used for various therapeutic
2012; Mihailovic et al., 2013; Nagalekshmi et al., 2011; indications but possess potential liver protection properties
Nithianantham et al., 2011; Paul et al., 2013; Shivananjappa stems from more than two decades of our research, as
et al., 2013; Singab et al., 2010; Mukherjee et al., 2009; reflected by our publications. In those articles, we looked for
DOI: 10.3109/13880209.2014.950387 Hepatoprotective properties of plant active constituents 783

possible underlying common mechanisms of cytoprotection also recognize that experimental pharmacological studies on
from molecular levels to whole animal studies of various medicinal plant constituents are being done, but clinical
structurally unrelated molecules. We found that several studies lack behind and need completion. Hence this review
compounds experimentally showed some hepato-ameliorative deals with constituents that are being tested experimentally
properties and enumerated possible explanations (Canova with or without clinical studies. They are arranged according
et al., 2007; Cerny et al., 2009; Farghali & Masek, 1998; to the wide use and the relevant experimental and/or clinical
Farghali et al., 1991a,b, 1994, 1996a,b, 1997, 2000b, 2002, trials in Table 1. For instance, silymarin obtained from the
2003; Gasbarrini et al., 1992a,b, 1993; Kmonickova et al., seeds of Silybum marianum (L.) Gaertner (Asteraceae) is the
2001; Lekic et al., 2011; Sakr et al., 1991a,b). most thoroughly investigated compound with potent anti-
hepatotoxic activity. Silymarin is a mixture of isomeric
Aim of the article flavolignans silybin, silydianin, and silychristen. It is
This review highlights the literature about purified or semi- important to study the hepatic effects of these pure substances
purified chemicals of herbal origins (i.e., chemically defined using in vitro and in vivo models, followed by genomic and
molecules) with reported experimental and/or clinical hepa- proteomic studies, and finally complemented with clinical
toprotective activity. The findings are likely to help further to evaluation for some of them. Here we give a simple table for
investigate hepatoprotective agents at both the experimental the most promising compounds that were tested at experi-
and the clinical levels. In addition to our personal experience, mental levels and some at clinical levels. In addition, recent
we have performed a literature search using mainly Web of advances in several natural products from plant origin for the
Science (WOS), PubMed, and Google search. An extensive potential treatment of several liver diseases were described as
number of articles which address many plant constituents with wogonin, naringenin, geniposide, rhein, and the list is
Downloaded by [202.67.40.194] at 09:32 06 October 2017

reported experimental (in vitro or in vivo) effects are increasing (Alvari et al., 2012; Zhang, 2013a).
summarized in various reviews (Alvari et al., 2012; Dhiman Next we describe in brief the most popular models in
et al., 2012; Ding et al., 2012; Girish & Pradhan, 2012; Muriel experimental testing of potential hepatoprotective com-
& Rivera-Espinoza, 2008; Zhang et al., 2013a). In this article, pounds. Drug constituents used for the treatment of viral
we only consider literature related to the most widely studied hepatitis that exhibit very favorable results are extensively
active plant constituents of chemically defined molecules reviewed elsewhere (Ahmed et al., 2008; Chen & Lai, 2013;
with potential hepatoprotective activity and the models most Thabrew & Hughes, 1996).
commonly used for their validation. We believe that these
In vitro and in vivo experimental hepatotoxic models
findings will certainly increase the likelihood of using
in liver research
hepatoprotective agents of well-defined molecules with less
adverse effects and will help to design new molecules by Successful development of therapy for the liver depends on
using computational and synthetic chemistry. Naturally, this the suitability of in vitro and in vivo test model systems for
requires examination of the molecular aspects of mode of hepatic injury. Several models are available to screen the
actions of these compounds. This may seem more intricate antihepatotoxic activity of any substance. Since there are
since both the impairment of the liver is multifactorial (viral limitations of the outcomes in each model, it is important to
or protozoal infections, chronic use of excessive alcohol, combine different methods for confirmation of the findings
drugs, xenobiotics, etc.) and the chemoprotective agents are (Muriel, 2008).
different in their chemical structures. However, there is an There are several in vitro models (e.g., hepatocyte cultures,
urgent need to find out a range of efficient drugs that may be perfused hepatocytes) that examine pathophysiological inju-
classified as hepatoprotective agents. In the literature, the use ries due to various chemicals (e.g., hepatotoxins, hypoxia or
of terms such as nutraceuticals, functional foods, herbal anoxia, anoxia/reoxygenation models in perfused immobi-
extracts, bioactive dietary constituents, phytochemicals, and lized hepatocytes) (Cerny et al., 2009; Farghali et al., 1991a,
similar terms is becoming widespread. 2000b; Gasbarrini et al., 1992b).
A number of in vivo models are depicted in Figure 1. Most
Potential beneficial plant constituents for of the studies investigating the hepatoprotective potential are
liver diseases of experimental nature. Some clinical studies were performed
using certain constituents (see later section); however, the
Natural products from plants used traditionally as hepatopro- majority of the studies are performed on herbal extracts or
tective have been reviewed by several authors (Adewusi & powdered dry plants for a specific liver disease.
Afolayan, 2010; Alvari et al., 2012; Dhiman et al., 2012; Ding
et al., 2012; Girish & Pradhan, 2012; Muriel & Rivera-
Are there possible common mechanisms of action in
Espinoza, 2008; Valan et al., 2010; Zhang et al., 2013a), who
hepatoprotection among various plant constituents?
described the phytoconstituents with hepatoprotective proper-
ties and classified them under phenyl compounds, coumarins, The answer is probably yes. In general, oxidative cell injury is
essential oils, monoterpenoids, diterpenoids, triterpenoids, well known to be associated both with the process of
steroids, alkaloids, and others. organisms aging and with numerous chronic diseases, such
In the present article, we focused on certain plant active as diabetes, atherosclerosis, macular degeneration, cataract,
constituents considered to be most promising/studied for liver and Alzheimers disease, among others. In addition, oxidative
diseases, and were critically investigated from the basic stress which is the cardinal mechanism that can be induced by
science point of view and, to some extent, the clinical one. We toxins and environmental factors leads to the accumulation of
784 H. Farghali et al. Pharm Biol, 2015; 53(6): 781791

Table 1. Most frequently studied hepatoprotective phytochemicals.

Compound Chemical class Major food resources Major biological activity References
Silymarin Mixture of flavonolignans Seeds of milk thistle Antioxidant, anti-inflammatory, Abenavoli et al. (2010), Ding et al.
(Silybin) consisting of among others Silybum marianum anti-fibrotic, protein synthesis (2012), El-Kamary et al. (2009),
of silibinin (7080%), isosi- increasing/regenerative, mem- Falasca et al. (2008), Farghali
libinin, silicristin, and brane stabilizing, and toxin et al. (2000a), Ferenci et al.
silidianin blocking activities; it reduces (1989), Gharagozloo et al.
tumor cell proliferation, angio- (2009), Glauert et al. (2010),
genesis as well as insulin Haddad et al. (2011), Loguercio
resistance et al. (2012), Mereish et al.
(1991), Pradhan and Girish
(2006), Salamone et al. (2012a, b),
Singh et al. (2012), Thabrew and
Huges (1996), Wagner et al.
(1968, 1974), and Zhang et al.
(2013a,b)
Curcumin Polyphenol: (1E,6E)-1,7-bis(4- Rhizomes turmeric Antioxidant, anti-inflammatory, Cerny et al. (2011), Ding et al.
hydroxy-3-methoxyphenyl)- (Curcuma longa), antifibrotic, anticancer, anti- (2012), Gupta et al. (2013),
1,6-heptadiene-3,5-dione yellow spice agreggatory, and potent cyto- Maiti et al. (2007), Kim et al.
chrome P450 inhibitory (2013), Singh et al. (2012),
activities Zhang et al. (2013a), Zhou et al.
(2011)
Quercetin Flavonol: 2-(3,4-dihydroxy- Fruits (apples, cranber- Antioxidant, anti-inflammatory, Chirumbolo (2010), Egert et al.
phenyl)-3,5,7-trihydroxy- ries), vegetables and anticancer activities (2009), Lekic et al. (2013),
Downloaded by [202.67.40.194] at 09:32 06 October 2017

4H-chromen-4-one (broccoli, onion), tea and Zhao et al. (2011)


leaves (Camellia
sinensis), grains
(buckwheat)
Resveratrol Polyphenol: trans-3,5,4- Berries, grapes, wine, Antioxidant, anti-inflammatory, Aftab et al. (2010), Athar et al.
trihydroxystilbene peanuts antiaging, antithrombotic, anti- (2009), Bishayee et al. (2010),
fibrogenic, and regenerative Cerny et al. (2009), Farghali
properties; it reduces tumor cell et al. (2009), Glauert et al.
proliferation, angiogenesis as (2010), Li et al. (2014), Patel
well as insulin resistance et al. (2011), Smoliga and
Rundell (2011), Smoliga et al.
(2012), Suchankova et al.
(2009), Timmers et al. (2012),
Wong et al. (2009), and Zhang
et al. (2013a)
Glycyrrhizin Triterpenic glycoside: Root of Indian licorice Antioxidant, anti-inflammatory, Jayaprakasam et al. (2009),
(3b,18a)-30-hydroxy-11,30- (Glycyrrhiza glabra) anticancer, antiviral, and immu- Ni et al. (2011), Orlent et al.
dioxoolean-12-en-3-yl 2-O- nomodulatory activities (2006), Wan et al. (2009), Yasui
b-D-glucopyranuronosyl- et al. (2011), Zhang et al.
b-D-glucopyranosid-uronic (2013a)
acid
Colchicine Tricyclic polymethoxy-deriv- Plants of the genus Anti-inflammatory, antifibrotic, Arrieta et al. (2006), Leung et al.
ate: N-[(7S)-1,2,3,10-tetra- Colchicum (autumn and immunomodulatory (2011), Muntoni et al. (2010),
methoxy-9-oxo-5,6,7,9-tetra- crocus, Colchicum activities and Nikolaidis et al. (2006)
hydrobenzo[a]heptalen-7- autumnale)
yl]acetamide

reactive oxygen/nitrogen species (ROS/RNS), further causing progression of fibrosis and that oxidative stress-related
imbalance in pro-oxidant/antioxidant steady state, known as molecules may act as mediators of molecular and cellular
oxidative stress. Protein cross-linking, lipid peroxidation, events implicated in liver fibrosis. Oxidative stress causes
mitochondrial dysfunction, and induction of cell death lipid peroxidation resulting in the formation of highly
pathways are among the identified mechanisms of cellular immunogenic molecules, damages proteins and DNA, induces
damage due to oxidative stress (Jaeschke, 2011; Jaeschke necro-apoptosis of hepatocytes, and amplifies the inflamma-
et al., 2012; Yeum et al., 2010). In this review, we discuss the tory response. ROS also stimulates the production of the pro-
role of ROS in liver fibrosis, a liver disease with significant fibrogenic mediator TGF-b from Kupffer cells and the
morbidity and mortality that affects 100 million people circulating inflammatory cells, as well as directly activates
worldwide. The role of oxidative stress in liver fibrosis, and hepatic stellate cells, resulting in the initiation of fibrosis.
pathways involved in hepatic fibrosis, are well covered in a Advances in understanding the mechanisms involved in
recent review article by Sanchez-Valle et al. (2012). ROS play fibrosis have identified new molecular targets with thera-
an important role in initiating hepatic fibrogenesis. Fibrosis, peutic potential for more targeted intervention of this disease.
manifested in several chronic liver diseases, is characterized In this regard, it is established that food rich in antioxidants,
by excessive collagen deposition in the extracellular matrix in such as vegetables and fruits, could reduce risk of several
response to activation of hepatic stellate cells. Clinical and chronic diseases. Therefore, it is reasonable to assume that
experimental data suggest that oxidative stress mediates the physiological doses of antioxidants that might be present in an
DOI: 10.3109/13880209.2014.950387 Hepatoprotective properties of plant active constituents 785
Downloaded by [202.67.40.194] at 09:32 06 October 2017

Figure 1. Common in vivo models of liver damage.

appropriate fruit and vegetable diet can establish an effective hepato-protective against steatosis and insulin resistance both
antioxidant network in vivo with consequent cytoprotective in vivo and in vitro, partly through regulating the IRS-1/PI3K/
effects on liver. Akt pathway which is involved in the pathogenesis of NAFLD
Various basic science literature contains enormous num- (Zhang et al., 2013b). Silibinin was also reported to improve
bers of molecules of plant origin that have the promise to hepatic oxidative stress and inflammation through the c-Jun
ameliorate diseases such as cancer to slowing the aging NH2-terminal kinase (Salamone et al., 2012a) and nuclear
process. However, most of these compounds were not factor kappa B (NF-kB) (Salamone et al., 2012b) pathways.
sufficiently evaluated in humans. A wide variety of plant In a clinical trial, silibinin given for 12 months in combin-
constituents, including silymarin, resveratrol, curcumin, quer- ation with vitamin E and phosphatidylcholine improved
cetin, and glycyrrhizin, have been reported to have multiple hepatic enzymes and insulin resistance in NAFLD patients
biological activities, mainly due to their antioxidant and anti- (Loguercio et al., 2012). The anti-inflammatory, immune-
inflammatory properties (Alvari et al., 2012; Dhiman et al., modulating activity, antifibrotic, and antioxidant effects of
2012; Ding et al., 2012; Girish & Pradhan, 2012; Muriel & silymarin that contribute to its hepatoprotective effect are
Rivera-Espinoza, 2008; Zhang et al., 2013a). The present summarized in a review by Abenavoli et al. (2010).
article focuses on only few well-studied naturally occurring Resveratrol, a polyphenolic compound with antioxidant
hepatoprotective (cytoprotective) compounds. properties, has been studied for the treatment of numerous
The seeds of milk thistle have been used for over 2000 pathologies, including NAFLD. Treatment with resveratrol
years for the treatment of liver diseases. Silybin, also known ameliorated NAFLD features, including insulin resistance,
as silibinin, is the major constituent (7080%) as well as the histological changes, glucose tolerance, inflammation, and
most active biological component of milk thistle. Laboratory oxidative stress, and restored IkBa (NF-kB inhibitor) which
studies revealed there is no real difference in activity between resulted in reduced activity of NF-kB (Li et al., 2014). Several
silymarin and silibinin in some experimental models. thousand articles deal with the biology and pharmacology of
Pharmacological and clinical studies showed that silymarin resveratrol, including many reports dealing with the molecu-
is relatively safe at the dosage range studied clinically (e.g., lar mechanisms of resveratrols cytoprotection (Suchankova
100150 mg daily in divided doses) (Ferenci et al., 1989). et al., 2009; Wong et al., 2009). The vast interest in
Silymarins protective effect against oxidative stress was resveratrol, which inspired an enormous number of studies,
partially attributed to a reduction in the intracellular calcium has led to the identification of multiple molecular targets of
in a model of perfused immobilized hepatocytes, resulting in resveratrol (Aftab et al., 2010; Athar et al., 2009; Bishayee
better functioning hepatocytes (Farghali et al., 2000b). The et al., 2010; Smoliga & Rundell 2011; Smoliga et al., 2012).
possible mechanism(s) that contribute to the hepatoprotective Resveratrol also acts as anti-inflammatory (Bereswill et al.,
effect of silymarin and the role played by intracellular 2010), alters drug metabolizing enzymes (Chow et al., 2010),
calcium was investigated using tert-butyl hydroperoxide and inhibits cyclooxygenases (Wendeburg et al., 2009), and,
D-galactosamine toxicity in the isolated immobilized and importantly, has specific effects on proteins and/or signaling
perfused hepatocytes model. Silibinin was also found to be cascades such as SIRT1 (silent information regulator T1)
786 H. Farghali et al. Pharm Biol, 2015; 53(6): 781791
Downloaded by [202.67.40.194] at 09:32 06 October 2017

Figure 2. The most important multiple pathways in the liver pathobiology that can be interrupted at one or more levels by hepatoprotective plant
constituents (not shown) to interfere with oxidative stress. ACC, acetyl-CoA carboxylase; AMPK, adenosine-50 -monophosphate-activated protein
kinase; ATP, adenosine triphosphate; Bax, proapoptotic protein of Bcl-2 family; BSEP, bile salt export pump; CAT, catalase; CO, carbon monoxide;
COX-2, inducible cyclooxygenase; CYP, cytochrome P450; eCa2+, extracellular calcium; ECM, extracellular matrix; eNOS (NOS-1), endothelial
nitric oxide synthase; ER, endoplasmic reticulum; GPx, glutathione peroxidase; GSH, reduced glutathione; GST, glutathione-S-transferase; HO-1,
inducible heme oxygenase; IL, interleukin; iCa2+, intracellular calcium; iNOS (NOS-2), inducible nitric oxide synthase; MDR3, multidrug resistance
protein 3; MRP2, multidrug resistance-related protein 2; NO, nitric oxide; NF-kB, nuclear factor kappa-B; NRS, nitrogen reactive species; ONOO,
peroxynitrite anion; p53, protein p53; PPAR-a/g, peroxisome proliferator-activated receptor-a/g; ROS, reactive oxygen species; SIRT1, silent
information regulator T1; SOD, superoxide dismutase; TGF-b, transforming growth factor-b; TNF-a, tumor necrosis factor-a, VCAM-1, vascular cell
adhesion molecule-1.

and AMPK (adenosine-50 -monophosphate-activated protein transcription factor. The fact that NF-kB has been associated
kinase) (Fullerton & Steinberg, 2010; Xiong et al., 2011) that with the induction of pro-inflammatory gene-expression
are summarized by Smoliga et al. (2012). The most studied makes research on agents which inhibit NF-kB an interesting
molecular mechanisms which might represent part of topic.
common pathway responsible for a hepatoprotective action Studies on resveratrol pretreatment effects on the synergy
that controls a number of regulatory factors associated between D-galactosamine (D-GalN) and LPS-induced liver
with metabolism, inflammation, and other pathophysiological failure in rats, and its effects on chemical prooxidants in
pathways, e.g., SIRT1 and AMPK are depicted in Figure 2. immobilized perfused hepatocytes was investigated in our
Resveratrol, a SIRT1 activator, increased AMPK phos- laboratory (Cerny et al., 2009; Farghali et al., 2009).
phorylation and reduced oxidative damage biomarkers during Parameters studied included liver function; plasma nitrite as
aging in F 2 hybrid mice (Suchankova et al., 2009). In a measure of nitric oxide (NO); non-enzymatic and enzymatic
addition, recent studies indicate intricate relationships among antioxidants in plasma and liver homogenate; and morpho-
resveratrol, nuclear factors, autacoids, and cytoprotection in logical examinations were performed using light and electron
various cells, tissues, or organs. For instance, in one study, microscopy. Observations related to pharmacological
resveratrol suppressed lipopolysaccharide (LPS)-induced increases of inducible nitric oxide synthase (NOS-2)/NO
nuclear translocation and activation of NF-kB thus inhibiting and inducible hemeoxygenase (HO-1)/carbon monoxide (CO)
pro-inflammatory responses in C6 microglia (Kim et al., in fulminant hepatic failure and modulation by resveratrol
2007). A similar finding about the protective effect of were followed up by real-time reverse transcription PCR (RT-
resveratrol as an inhibitor of NF-kB-mediated vascular cell PCR) in liver tissue. In the last study, we found that reduction
adhesion molecule (VCAM-1) induction was reported in NO production, down-regulation of NOS-2 expression,
(Carluccio et al., 2007). Recently, NF-kB was suggested as modification of oxidative stress parameters, and modulation
a target for drug therapy in liver diseases where resveratrol of HO-1 are among the mechanisms responsible for the
was among several agents that inhibits the NF-kB cytoprotective effect of resveratrol in the LPS/D-GalN liver
DOI: 10.3109/13880209.2014.950387 Hepatoprotective properties of plant active constituents 787

toxicity, and in tert-butylhyroperoxide-induced hepatocyte resistance, and liver histology, without increases in body
toxicity models. Resveratrol pretreatment led to the overall weight (Loguercio et al., 2012).
improvement in hepatotoxic markers and morphology after Although the present review is not dealing with antiviral
the hepatic insult. Several studies have also highlighted the agents for liver infection as mentioned before, we cite the
hepatoprotective properties of resveratrol (Bishayee et al., example of silybinvitamin Ephospholipid complex treat-
2010). ment in patients with HCV (Falasca et al., 2008). The treated
SIRT1 plays a cardinal role in the mode of action of group of HCV patients showed an improvement in hepatic
resveratrol through AMPK activity as shown in Figure 2. indices and viral load, and had a significant and persistent
Moghe et al. (2011) presented a relationship between histone reduction of ALT and AST serum levels. In this group,
modification and alcohol-induced liver disease with a focus cytokines showed a statistically significant modulation of
on epigenetic changes, including histone modifications that IL-2 and IL-6. After the treatment, the hepatic steatosis group
regulate gene expression during pathogenesis. As reported, showed a significant decrease in ALT, AST, g-GT, alkaline
chronic alcohol consumption also sensitizes non-immune phosphatase, total cholesterol, fasting glucose, insulinemia,
cells such as hepatocytes, in addition to immune cells, to HOMA (the homeostatic model assessment) value, and
inflammatory signals and impairs their ability to respond C-reactive protein. There was a significant reduction of
to protective signals. Based on these advances, a number of IFN-g, TNF-a, and IL-6. The data suggest that the complex
inflammatory targets have been identified with the potential exerts hepatoprotective, anti-inflammatory, and antifibrotic
for therapeutic intervention in alcoholic liver disease by using effects. The authors suggested that this treatment may,
such compounds, presenting new opportunities, and chal- therefore, be useful in clinical practice in patients with chronic
lenges for translational research (Wang et al., 2012; Zakhari & hepatitis C who cannot undergo conventional antiviral therapy.
Downloaded by [202.67.40.194] at 09:32 06 October 2017

Li, 2007). In a double-blind clinical trial, the combined therapy of


Oxidative stress and liver inflammation are also linked to silymarin and desferrioxamine in patients with b-thalasemia
diet-induced obesity, thus emphasizing the assumption that (iron-loaded patients) was beneficial (Gharagozloo et al.,
hepatoprotectors should have common pathways at certain 2009). Because of the poor absorption of silymarin, critical
level(s) (Peng et al., 2012). At this juncture, it is important to reassessment of its bioavailability (using an array of methods
emphasize the importance of SIRT1 allosteric modulators and that can improve its bioavailability, including complexion
cytoprotection including hepatoprotection. with b-cyclodextrins, solid dispersion method, formation of
To summarize, Figure 2 demonstrates that multipathways microparticles and nanoparticles, self-microemulsifying drug
in the liver pathobiology that can be interrupted at one or delivery systems, micelles, liposomes, and phytosomes) were
more levels, practically, by all natural hepatoprotective published (Javed et al., 2011). Even a sensitive LC-MS/MS
studied leading to interference with the oxidative stress. assay for the simultaneous analysis of the major active
components of silymarin in human plasma has been
developed to ensure regularity of plasma level of this agent
Importance of and emphasis on controlled clinical
(Brinda et al., 2012).
trials on active plant constituents
Resveratrol also received widespread attention as either a
Most clinical trials were conducted on herbal powders or potential therapeutic or a preventative agent for several
extracts, and a relatively low number of studies used pure diseases, which resulted in thousands of reports that described
active constituents. In spite of the several thousand reports the effects of resveratrol administration on physiological
that deal with favorable effects on various liver injury models, responses in animals and humans. The limited numbers of
clinical trials are lagging behind and relatively small numbers human clinical trials available have largely described various
were done on active constituents (Akachi et al., 2010; Kim aspects of resveratrols safety and bioavailability, reaching a
et al., 2013; Panahi et al., 2012; Sarwar et al., 2011; Singh & consensus that it is generally well tolerated, but poor
Sharma, 2011; Wong et al., 2013). bioavailability. The relatively recent GlaxoSmithKlines
The safety and efficacy of silymarin on signs and phase IIa study of SRT501 (a resveratrol pharmaceutical
symptoms, and biomarkers were investigated in randomized formulation in relatively high dose of resveratrol 5 g of
controlled clinical trials on patients with acute hepatitis. The SRT501) in advanced multiple myeloma has been concluded.
standard dose of silymarin (140 mg) produced earlier It was explained by the manufacturers that SRT501 offers
improvement in subjective and clinical markers of biliary minimal efficacy while having a potential to indirectly
excretion. With a modest sample size, and multiple etiologies exacerbate a renal complication common in the patient
for acute clinical hepatitis, the results suggest that standard population with multiple myeloma. Therefore, researchers are
recommended doses of silymarin are safe and may be hoping to find a way to concentrate the effect into a safe dose
potentially effective in improving symptoms of acute clinical within an effective therapeutic range (Smoliga et al., 2012).
hepatitis (El-Kamary et al., 2009). Given the worldwide increase in age-related metabolic
A randomized clinical trial was published on silybin diseases, the beneficial effects of resveratrol on metabolism
combined with phosphatidylcholine and vitamin E in patients and healthy aging in humans are currently a topic of intense
with non-alcoholic fatty liver disease. A preparation with the investigation. In fact, the occurrence of mild-to-moderate side
name of Realsil (RA) which comprises the silybin phytosome effects is likely to limit the doses employed in future trials
complex (silybin plus phosphatidylcholine) co-formulated to significantly less than 5 g/d and additional information is
with vitamin E was tested. Treatment for 12 months was needed to increase the chances of success in future clinical
associated with improvement in liver enzymes, insulin trials (Patel et al., 2011; Timmers et al., 2012).
788 H. Farghali et al. Pharm Biol, 2015; 53(6): 781791

Other molecules such as curcumin, quercetin, glycerhyzin, Aftab N, Likhitwitayawuid K, Vieira A. (2010). Comparative antioxidant
activities and synergism of resveratrol and oxyresveratrol. Nat Prod
or colchicine were clinically investigated in liver diseases. Res 24:172633.
According to recent report, more than 65 human clinical trials Ahmed MS, Wong CF, Shawki H, et al. (2008). Rapidly deteriorating
of curcumin, which included more than 1000 patients, have renal function with membranoproliferative glomerulonephritis type 1
been completed, and as many as 35 clinical trials are associated with hepatitis C treated successfully with steroids and
antiviral therapy: A case report and review of literature. Clin Nephrol
underway (Gupta et al., 2013). Curcumin was suggested to 69:298301.
counteract the adverse hepatotoxic side effects of anti- Akachi T, Shiina Y, Ohishi Y, et al. (2010). Hepatoprotective effects of
tuberculosis drug combinations, a possible valuable hepato- flavonoids from shekwasha (Citrus depressa) against D-galactosa-
protective application (Singh et al., 2012). In fact, clinical mine-induced liver injury in rats. J Nutr Sci Vitaminol 56:607.
Alonso FT, Garmendia ML, de Aguirre M, Searle J. (2010). Mortality
trials are underway using these plant constituents for various trend from liver cirrhosis in Chile from 1990 to 2007. Rev Med Chile
health applications including liver diseases (Del Prete et al., 138:12538.
2012; Egert et al., 2009). Clinical trials on licorice Alvari A, Mehrnaz SO, Ahmad FJ, Abdin MZ. (2012). Contemporary
or glycyrrhizin are increasing specifically in viral hepatitis overview on clinical trials and future prospects of hepato-protective
herbal medicines. Rev Recent Clin Trials 7:21423.
adjuvant therapy and autoimmune hepatitis (Orlent et al., Ansari JA, Ali S, Ansari MA. (2011). A brief focus on hepatoprotective
2006; Yasui et al., 2011). leads from herbal origin. Int J Pharmacol 7:21216.
Colchicine is a well-established drug for its anti-inflam- Arrieta O, Rodriguez-Diaz JL, Rosas-Camargo V, et al. (2006).
matory actions and is used as a medication for several Colchicine delays the development of hepatocellular carcinoma in
patients with hepatitis virus-related liver cirrhosis. Cancer 107:
diseases. It is effective clinically with ursodiol in a subset of 18528.
patients with primary biliary cirrhosis and was found to be an Asgarpanah J, Kazemivash N. (2012). Phytochemistry, pharmacology
effective and safe antifibrotic drug for long-term treatment of and medicinal properties of Coriandrum sativum L. Afr J Pharm
Pharmacol 6:23405.
Downloaded by [202.67.40.194] at 09:32 06 October 2017

chronic liver disease in which fibrosis progresses towards


Asuku O, Atawodi SE, Onyike E. (2012). Antioxidant, hepatoprotective,
cirrhosis (Leung et al., 2011; Muntoni et al., 2010). and ameliorative effects of methanolic extract of leaves of Grewia
mollis Juss. on carbon tetrachloride-treated Albino rats. J Med Food
15:838.
Conclusion Athar M, Back JH, Kopelovich L, et al. (2009). Multiple molecular
This article examines the current state-of-knowledge of the targets of resveratrol: Anti-carcinogenic mechanisms. Arch Biochem
Biophys 486:95102.
effects of pure natural plant constituents and possible Avila DS, Palma AS, Colle D, et al. (2011). Hepatoprotective activity
derivatives and their potential applications to human liver of a vinylic telluride against acute exposure to acetaminophen. Eur J
diseases. It utilizes this information to develop further Pharmacol 661:92101.
guidance for encouraging human clinical trials. From various Bereswill S, Munoz M, Fischer A, et al. (2010). Anti-inflammatory
effects of resveratrol, curcumin and simvastatin in acute small
literature and our own studies, it seems that all these active intestinal inflammation. Plos One [Online]. Available from: http://
herbal constituents act at some point to reduce ROS/RNS, www.plosone.org/article/info%3Adoi%2F10.1371%2Fjournal.pone.
leading to the hepato-ameliorative effect. Due to insufficient 0015099 [last accessed 23 Jun 2014].
scientific-based pharmacological data, most of the herbal Bilgi N, Bell K, Ananthakrishnan AN, Atallah E. (2010). Imatinib and
Panax ginseng: A potential interaction resulting in liver toxicity. Ann
formulations containing these molecules cannot be recom- Pharmacother 44:9268.
mended for the treatment of liver diseases. Hence, this review Bishayee A, Darvesh AS, Politis T, McGory R. (2010). Resveratrol and
article is intended to focus briefly on few herbal hepatopro- liver disease: From bench to bedside and community. Liver Int 30:
tective active constituents that may be useful to health 110314.
Blachier M, Leleu H, Peck-Radosavljevic M, et al. (2013). The burden of
professionals, scientists working in the field of pharmacology liver disease in Europe: A review of available epidemiological data.
and therapeutics to develop evidence-based hepatoprotective J Hepatol 58:593608.
agents. This can be accomplished by research on pure Brinda BJ, Zhu HJ, Markowitz JS. (2012). A sensitive LC-MS/MS assay
chemical structures derived from plants or after structural for the simultaneous analysis of the major active components of
silymarin in human plasma. J Chromatogr B 902:19.
modifications using computational chemistry to prepare new Canova NK, Kmonickova E, Martinek J, et al. (2007). Thapsigargin,
lead compounds. a selective inhibitor of sarco-endoplasmic reticulum Ca2+-ATPases,
The future is to carry out controlled prospective double- modulates nitric oxide production and cell death of primary rat
blind multicenter studies with the isolated active constituents hepatocytes in culture. Cell Biol Toxicol 23:33754.
Carluccio MA, Ancora MA, Massaro M, et al. (2007). Homocysteine
or related newly synthesized drugs with proven beneficial pre- induces VCAM-1 gene expression through NF-kappa B and NAD(P)H
clinical in vitro and in vivo effects where basic hepatobiology oxidase activation: Protective role of Mediterranean diet polyphenolic
should also be encouraged. antioxidants. Am J Physiol-Heart C 293:H234454.
Cerny D, Canova NK, Martinek J, et al. (2009). Effects of resveratrol
pretreatment on tert-butylhydroperoxide induced hepatocyte toxicity
Declaration of interest in immobilized perifused hepatocytes: Involvement of inducible
nitric oxide synthase and hemoxygenase-1. Nitric Oxide-Biol Ch 20:
This work was fully supported by the Charles University 18.
institutional program PRVOUK-P25/LF1/2. Cerny D, Lekic N, Vanova K, et al. (2011). Hepatoprotective effect of
curcumin in lipopolysaccharide/D-galactosamine model of liver injury
in rats: Relationship to HO-1/CO antioxidant system. Fitoterapia 82:
References 78691.
Chattopadhyay RR. (2003). Possible mechanism of hepatoprotective
Abenavoli L, Capasso R, Milic N, Capasso F. (2010). Milk thistle in liver activity of Azadirachta indica leaf extract: Part II. J Ethnopharmacol
disease: Past, present, future. Phytother Res 24:142332. 89:21719.
Adewusi EA, Afolayan AJ. (2010). A review of natural products with Chen Q, Lai HF. (2013). Plant-derived virus-like particles as vaccines.
hepatoprotective activity. J Med Plant Res 4:131834. Hum Vacc Immunother 9:3255.
DOI: 10.3109/13880209.2014.950387 Hepatoprotective properties of plant active constituents 789
Chirumbolo S. (2010). The role of quercetin, flavonols and flavones in Gasbarrini A, Borle AB, Caraceni P, et al. (1992b). Hepatocytes anoxic
modulating inflammatory cell function. Inflamm Allergy-Drug Targets injury is prevented by high concentration of fructose. Transpl P 24:
9:26385. 281012.
Chow HHS, Garland LL, Hsu CH, et al. (2010). Resveratrol modulates Gasbarrini A, Borle AB, Farghali H, et al. (1993). Fasting enhances the
drug- and carcinogen-metabolizing enzymes in a healthy volunteer effect of anoxia on ATP Ca2+ and cell injury in isolated rat
study. Cancer Prev Res 3:116875. hepatocytes. Biochim Biophys Acta 1178:919.
Del Prete A, Scalera A, Iadevaia MD, et al. (2012). Herbal products: Gharagozloo M, Moayedi B, Zakerinia M, et al. (2009). Combined
Benefits, limits, and applications in chronic liver disease. Evid-Based therapy of silymarin and desferrioxamine in patients with beta-
Compl Alt [Online]. Available from: http://www.hindawi.com/ thalassemia major: A randomized double-blind clinical trial. Fund
journals/ecam/2012/837939/ [last accessed 23 Jun 2014]. Clin Pharmacol 23:35965.
Dhiman A, Nanda A, Ahmad S. (2012). A recent update in research on Ghosh N, Ghosh R, Mandal V, Mandal SC. (2011). Recent advances in
the antihepatotoxic potential of medicinal plants. J Chinese Integr herbal medicine for treatment of liver diseases. Pharm Biol 49:
Med 10:11727. 97088.
Ding RB, Tian K, Huang LL, et al. (2012). Herbal medicines for the Girish C, Pradhan SC. (2012). Indian herbal medicines in the treatment
prevention of alcoholic liver disease: A review. J Ethnopharmacol of liver diseases: Problems and promises. Fund Clin Pharmacol 26:
144:45765. 1809.
Egert S, Bosy-Westphal A, Seiberl J, et al. (2009). Quercetin reduces Glauert HP, Calfee-Mason K, Stemm DN, et al. (2010). Dietary
systolic blood pressure and plasma oxidised low-density lipoprotein antioxidants in the prevention of hepatocarcinogenesis: A review.
concentrations in overweight subjects with a high-cardiovascular Mol Nutr Food Res 54:87596.
disease risk phenotype: A double-blinded, placebo-controlled cross- Gupta SS. (1994). Prospects and perspectives of natural plant products in
over study. Br J Nutr 102:106574. medicine. Indian J Pharmacol 26:112.
El-Kamary SS, Shardell MD, Abdel-Hamid M, et al. (2009). A Gupta SC, Kismali G, Aggarwal BB. (2013). Curcumin, a component of
randomized controlled trial to assess the safety and efficacy of turmeric: From farm to pharmacy. Biofactors 39:213.
silymarin on symptoms, signs and biomarkers of acute hepatitis. Gutierrez RMP, Gonzalez AMN, Hoyo-Vadillo C. (2013). Medicinal
Phytomedicine 16:391400. flowers. XXIV. Chemical structures and hepatoprotective effects of
Falasca K, Ucciferri C, Mancino P, et al. (2008). Treatment with silybin- constituents from flowers of Hedychium coronarium alkaloids from
Downloaded by [202.67.40.194] at 09:32 06 October 2017

vitamin E-phospholipid complex in patients with hepatitis C infection. piper: A review of its phytochemistry and pharmacology. Mini-Rev
J Med Virol 80:19006. Med Chem 13:16393.
Farghali H, Sakr M, Gasbarrini A, et al. (1991a). Effect of FK 506 Haddad Y, Vallerand D, Brault A, Haddad PS. (2011). Antioxidant and
chronic administration on bromsulphthalein hepatic excretion in rats. hepatoprotective effects of silibinin in a rat model of nonalcoholic
Transpl P 23:28024. steatohepatitis. Evid-based Complement Altern Med [Online].
Farghali H, Gasbarrini A, Borle AB, et al. (1991b). FK 506 modulates Available from: http://www.hindawi.com/journals/ecam/2011/647903/
D-galactosamine-induced hepatitis in rats. Transpl P 23:280911. [last accessed 23 Jun 2014].
Farghali H, Rilo H, Zhang WG, et al. (1994). Liver regeneration after Ho WY, Yeap SK, Ho CL, et al. (2012). Hepatoprotective activity of
partial hepatectomy in the rat: Sequential events monitored by P-31- Elephantopus scaber on alcohol-induced liver damage in mice. Evid-
nuclear magnetic resonance spectroscopy and biochemical studies. based Complement Altern Med [Online]. Available from: http://
Lab Invest 70:41825. www.hindawi.com/journals/ecam/2012/417953/ [last accessed 23 Jun
Farghali H, Bencko V, Kamenikova L, Hynie S. (1996a). Response of 2014].
immobilized hepatocytes in a perfusion system to anoxia/rexygena- Hong JY, Lebofsky M, Farhood A, Jaeschke H. (2009). Oxidant stress-
tion: Effect of cyclosporine A pretreatment. Physiol Res 45:227333. induced liver injury in vivo: Role of apoptosis, oncotic necrosis, and
Farghali H, Martinek J, Kamenikova L, Hynie H. (1996b). Amelioration c-Jun NH2-terminal kinase activation. Am J Physiol Gastrointest
of chemically-induced hepatocytes injury by cyclosporine A. Liver Physiol 296:G57281.
Pharmacol Res 34:21118. Jaeschke H. (2011). Reactive oxygen and mechanisms of inflammatory
Farghali H, Zidek Z, Hynie S. (1997). Effects of nitroprusside as a nitric liver injury: Present concepts. J Gastroenterol Hepatol 26:1739.
oxide donor on anoxia/reoxygenation and D-galactosamine hepatic Jaeschke H, McGill MR, Ramachandran A. (2012). Oxidant stress,
injuries: A study in perfused hepatocytes. Physiol Res 46:3639. mitochondria, and cell death mechanisms in drug-induced liver injury:
Farghali H, Masek K. (1998). Immunopharmacologic agents in the Lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev
amelioration of hepatic injuries. Int J Immuinopharmacol 20:12539. 44:88106.
Farghali H, Kamenikova L, Hynie S, Kmonickova E. (2000a). Silymarin Javed S, Kohli K, Ali M. (2011). Reassessing bioavailability of
effects on intracellular calcium and cytotoxicity: A study in perfused silymarin. Altern Med Rev 16:23949.
rat hepatocytes after oxidative stress injury. Pharmacol Res 41:2317. Jayaprakasam B, Doddaga S, Wang R, et al. (2009). Licorice flavonoids
Farghali H, Kmonickova E, Lotkova H, Martinek J. (2000b). Evaluation inhibit eotaxin-1 secretion by human fetal lung fibroblasts in vitro.
of calcium channel blockers as potential hepatoprotective agents in J Agric Food Chem 57:8205.
oxidative stress injury of perfused hepatocytes. Physiol Res 49:2618. Khan S, Khan I, Walker L. (2008). Botanical supplement and hepato-
Farghali H, Canova N, Gaier N, et al. (2002). Inhibition of endotoxemia- toxicity. In: Smart RC, Hodgson E, eds. Molecular and Biochemical
induced nitric oxide synthase expression by cyclosporin A enhances Toxicology, 4th ed. USA: John Wiley & Sons Inc, 591606.
hepatocyte injury in rats: Amelioration by NO donors. Int Kim YA, Kim GY, Park KY, Choi YH. (2007). Resveratrol inhibits nitric
Immunopharmacol 2:11727. oxide and prostaglandin E-2 production by lipopolysaccharide-
Farghali H, Canova N, Kucera T, et al. (2003). Nitric oxide synthase activated C6 microglia. J Med Food 10:21824.
inhibitors modulate lipopolysaccharide-induced hepatocyte injury: Kim SW, Ha KC, Choi EK, et al. (2013). The effectiveness of fermented
Dissociation between in vivo and in vitro effect. Int Immunopharmacol turmeric powder in subjects with elevated alanine transaminase levels:
3:162738. A randomised controlled study. BMC Complem Altern M [Online].
Farghali H, Cerny D, Kamenikova L, et al. (2009). Resveratrol attenuates Available from: http://www.biomedcentral.com/1472-6882/13/58 [last
lipopolysaccharide-induced hepatitis in D-galactosamine sensitized accessed 23 Jun 2014].
rats: Role of nitric oxide synthase 2 and heme oxygenase-1. Nitric Kmonickova E, Drahota Z, Kamenikova L, et al. (2001). Modulatory
Oxide-Biol Ch 21:21625. effect of cyclosporin A on tert-butyl hydroperoxide-induced oxida-
Ferenci P, Dragosics B, Dittrich H, et al. (1989). Randomized controlled tive damage in hepatocytes. Immunopharmacol Immunotoxicol 23:
trial of silymarin treatment in patients with cirrhosis of the liver. 4354.
J Hepatol 9:10513. Lee BH, Huang YY, Duh PD, Wu SC. (2012). Hepatoprotection of
Fullerton MD, Steinberg GR. (2010). SIRT1 takes a backseat to AMPK emodin and Polygonum multiflorum against CCl4-induced liver injury.
in the regulation of insulin sensitivity by resveratrol. Diabetes 59: Pharm Biol 50:3519.
5513. Lekic N, Cerny D, Horinek A, et al. (2011). Differential oxidative stress
Gasbarrini A, Borle AB, Farghali H, et al. (1992a). Effect of anoxia on responses to D-galactosamine-lipopolysaccharide hepatotoxicity based
intracellular ATP, Na+, Cai2+ and cytotoxicity in rat hepatocytes. on real time PCR analysis of selected oxidant/antioxidant and
J Biol Chem 267:665463. apoptotic gene expressions in rat. Physiol Res 60:54958.
790 H. Farghali et al. Pharm Biol, 2015; 53(6): 781791

Lekic N, Canova NK, Horinek A, Farghali H. (2013). The involvement Pradhan SC, Girish C. (2006). Hepatoprotective herbal drug, silymarin
of heme oxygenase 1 but not nitric oxide synthase 2 in a from experimental pharmacology to clinical medicine. Indian J Med
hepatoprotective action of quercetin in lipopolysaccharide-induced Res 124:491504.
hepatotoxicity of D-galactosamine sensitized rats. Fitoterapia 87: Rana SV, Pal R, Vaiphei K, et al. (2011). Garlic in health and disease.
206. Nutr Res Rev 24:6071.
Leung J, Bonis PA, Kaplan MM. (2011). Colchicine or methotrexate, Rayfield B. (2013). Why we need extra liver protection. Sinclair Method
with ursodiol, are effective after 20 years in a subset of patients with [Online]. Available from: http://www.sinclairmethod.com/index_files/
primary biliary cirrhosis. Clin Gastroenterol H 9:77680. Page563.htm [last accessed 25 Feb 2014].
Li L, Hai J, Li Z, et al. (2014). Resveratrol modulates autophagy and Sakr MF, Zetti GM, Hassanein TH, et al. (1991a). Van Thiel, DH: FK
NF-kB activity in murine model for treating non-alcoholic fatty liver 506 ameliorates the hepatic injury associated with ischemia and
disease. Food Chem Toxicol 63:16673. reperfusion in rats. Hepatology 13:94751.
Loguercio C, Andreone P, Brisc C, et al. (2012). Silybin combined with Sakr MF, Zetti GM, Farghali H, et al. (1991b). Protective effect of FK
phosphatidylcholine and vitamin E in patients with nonalcoholic fatty 506 against hepatic ischemia in rats. Transpl P 23:3401.
liver disease: A randomized controlled trial. Free Radical Bio Med 52: Salamone F, Galvano F, Marino A, et al. (2012a). Silibinin improves
165865. hepatic and myocardial injury in mice with non-alcoholic steatohe-
Maiti K, Mukherjee K, Gantait A, et al. (2007). Curcumin-phospholipid patitis. Digest Liver Dis 44:33442.
complex: Preparation, therapeutic evaluation and pharmacokinetic Salamone F, Galvano F, Cappello F, et al. (2012b). Silibinin modulates
study in rats. Int J Pharm 330:15563. lipid homeostasis and inhibits nuclear factor kappa B activation in
Mereish KA, Bunner DL, Ragland DR, Creasia DA. (1991). Protection experimental non-alcoholic steatohepatitis. Transl Res 159:47786.
against microcystin-LR-induced hepatotoxicity by silymarin: Sanchez-Valle V, Chavez-Tapia NC, Uribe M, Mendez-Sanchez N.
Biochemistry, histopathology, and lethality. Pharm Res 8:2737. (2012). Role of oxidative stress and molecular changes in liver
Mihailovic V, Mihailovic M, Uskokovic A, et al. (2013). fibrosis: A review. Curr Med Chem 19:485060.
Hepatoprotective effects of Gentiana asclepiadea L. extracts against Sarwar M, Attitalla IH, Abdollahi M. (2011). A review on the recent
carbon tetrachloride induced liver injury in rats. Food Chem Toxicol advances in pharmacological studies on medicinal plants: Animal
52:8390. studies are done but clinical studies needs completing. Asian J Anim
Moghe A, Joshi-Barve S, Ghare S, et al. (2011). Histone modifications Vet Adv 6:86783.
Downloaded by [202.67.40.194] at 09:32 06 October 2017

and alcohol-induced liver disease: Are altered nutrients the missing Shivananjappa MM, Mhasavade D, Joshi MK. (2013). Aqueous extract
link? World J Gastroenterol 17:246572 of Terminalia arjuna attenuates tert-butyl hydroperoxide-induced
Mukherjee PK, Sahoo AK, Narayanan N, et al. (2009). Lead finding oxidative stress in HepG2 cell model. Cell Biochem Funct 31:12935.
from medicinal plants with hepatoprotective potentials. Expert Opin Singab ANB, Ayoub NA, Ali EN, Mostafa NM. (2010). Antioxidant and
Drug Dis 4:54576. hepatoprotective activities of Egyptian moraceous plants against
Muntoni S, Rojkind M, Muntoni S. (2010). Colchicine reduces carbon tetrachloride-induced oxidative stress and liver damage in rats.
procollagen III and increases pseudocholinesterase in chronic liver Pharm Biol 48:125564.
disease. World J Gastroenterol 16:288994. Singh H, Sharma YK. (2011). Clinical evaluation of the hepatoprotective
Muriel P. (2008). Some experimental models in liver damage. In: Sahu effect of Katuki (Picrorhiza kurroa Royle ex Benth.) processed in
SC, ed. Hepatotoxicity: From Genomics to In Vitro and In Vivo Guduchi (Tinospora cordifolia Wild.) Miers in patients receiving lipid
Models. 1st ed. London, UK: John Wiley & Sons Inc., 11937. lowering drugs (statins). Indian J Tradit Know 10:65760.
Muriel P, Rivera-Espinoza Y. (2008). Beneficial drugs for liver diseases. Singh M, Sasi P, Gupta VH, et al. (2012). Protective effect of curcumin,
J Appl Toxicol 28:93103. silymarin and N-acetylcysteine on antitubercular drug-induced
Nagalekshmi R, Menon A, Chandrasekharan DK, Nair CKK. (2011). hepatotoxicity assessed in an in vitro model. Hum Exp Toxicol 31:
Hepatoprotective activity of Andrographis paniculata and Swertia 78897.
chirayita. Food Chem Toxicol 49:336773. Smoliga JM, Rundell KW. (2011). Oxidative stress: Friend and foe of the
National End of Life Care Intelligence Network (NEoLCIN). (2013). elite marathoner. J Appl Physiol 110:2877.
Deaths from liver disease: Implications for end of life care in England. Smoliga JM, Vang O, Baur JA. (2012). Challenges of translating basic
Public Health England [Online]. Available from: http://www. research into therapeutics: Resveratrol as an example. J Gerontol
endoflifecare-intelligence.org.uk/resources/publications/deaths_from_ A-Biol 67:15867.
liver_disease [last accessed 25 Feb 2014]. Subramanian R, Asmawi MZ, Sadikun A. (2012). A bitter plant with a
Ni YF, Kuai JK, Lu ZF, et al. (2011). Glycyrrhizin treatment is sweet future? A comprehensive review of an oriental medicinal plant:
associated with attenuation of lipopolysaccharide-induced acute lung Andrographis paniculata. Phytochem Rev 11:3975.
injury by inhibiting cyclooxygenase-2 and inducible nitric oxide Suchankova G, Nelson LE, Gerhart-Hines Z, et al. (2009). Concurrent
synthase expression. J Surg Res 165:E2935. regulation of AMP-activated protein kinase and SIRT1 in mammalian
Nikolaidis N, Kountouras J, Giouleme O, et al. (2006). Colchicine cells. Biochem Biophys Res Comman 378:83641.
treatment of liver fibrosis. Hepato-Gastroenterol 53:2815. Thabrew MI, Hughes RD. (1996). Phytogenic agents in the therapy of
Nithianantham K, Shyamala M, Chen Y, et al. (2011). Hepatoprotective liver disease. Phytother Res 10:4617.
potential of Clitoria ternatea leaf extract against paracetamol induced Timmers S, Auwerx J, Schrauwen P. (2012). The journey of resveratrol
damage in mice. Molecules 16:1013445. from yeast to human. Aging-US 4:14658.
Nobili V, Carter-Kent C, Feldstein AE. (2011). The role of lifestyle Torres DM, Williams CD, Harrison SA. (2012). Features, diagnosis, and
changes in the management of chronic liver disease. BMC Med. treatment of nonalcoholic fatty liver disease. Clin Gastroenterol
[Online], 9, Article Number: 70 [Epub ahead of print]. doi:10.1186/ Hepatol 10:83758.
1741-7015-9-70. Torres-Gonalez L, Munoz-Espinosa LE, Rivas-Estilla AM, et al. (2011).
Orlent H, Hansen BE, Willems M, et al. (2006). Biochemical and Protective effect of four Mexican plants against CCl4-induced damage
histological effects of 26 weeks of glycyrrhizin treatment in chronic on the Huh7 human hepatoma cell line. Ann Hepatol 10:739.
hepatitis C: A randomized phase II trial. J Hepatol 45:53946. Upadhyay R, Tiwari KN, Singh K. (2013). High frequency shoots
Panahi Y, Ghamarchehreh ME, Beiraghdar F, et al. (2012). Investigation regeneration for mass multiplication of Phyllanthus fraternus Webster
of the effects of Chlorella vulgaris supplementation in patients with An important antiviral and hepatoprotective plant. Appl Biochem
non-alcoholic fatty liver disease: A randomized clinical trial. Hepato- Biotechnol 169:230314.
Gastroenterol 59:2099103. Valan MF, Britto AJB, Venkataraman R. (2010). Phytoconstituens with
Patel KR, Scott E, Brown VA, et al. (2011). Clinical trials of resveratrol. hepatoprotective activity. Int J Chem Sci 8:142132.
Ann NY Acad Sci 1215 (Resveratrol and Health):1619. Wagner H, Horhammer L, Munster R. (1968). The chemistry of silymarin
Paul S, Hwang JK, Kim HY, et al. (2013). Multiple biological properties (silybin), the active principle of the fruits of Silybum marianum (L.)
of macelignan and its pharmacological implications. Arch Pharm Res Gaertn. (Carduus marianus) (L.). Aarzneimittelfoschung/Drug Res 18:
36:26472. 68896.
Peng YH, Rideout D, Rakita S, et al. (2012). Source: Diet-induced Wagner H, Diesel P, Seitz M. (1974). Chemistry and analysis of
obesity associated with steatosis, oxidative stress, and inflammation in silymarin from Silybum marianum (L.). Arzneimittelforschung/Drug
liver. Surg Obes Relat Dis 8:7381. Res 24:46671.
DOI: 10.3109/13880209.2014.950387 Hepatoprotective properties of plant active constituents 791
Wan XY, Luo M, Li XD, He P. (2009). Hepatoprotective and anti- Yeum KJ, Russel RM, Aldini G. (2010). Biomarkers for reassemble the
hepatocarcinogenic effects of glycyrrhizin and matrine. Chem-Biol results and re-create the whole by human studies. In: Aldini G, Yeum
Interact 181:1519. KJ, Niki E, Russel RM, eds. Biomarkers for Antioxidant Defence and
Wang HJ, Gao B, Zakhari S, Nagy LE. (2012). Inflammation in Oxidative Damage: Principles and Practical Applications. Ames,
alcoholic liver disease. Ann Rev Nutr 32:34368. Iowa: Blackwell Publishing, 319.
Wendeburg L, de Oliveira ACP, Bhatia HS, et al. (2009). Resveratrol Zakhari S, Li TK. (2007). Determinants of alcohol use and abuse: Impact
inhibits prostaglandin formation in IL-1 beta-stimulated SK-N-SH of quantity and frequency patterns on liver disease. Hepatology 46:
neuronal cells. J Neuroinflamm. [Online], 6, Article Number: 26, 20329.
[Epub ahead of print]. doi:10.1186/1742-2094-6-26. Zhang H, Yu CH, Jiang YP, et al. (2012). Protective effects of polydatin
Wong YT, Gruber J, Jenner AM, et al. (2009). Elevation of oxidative- from Polygonum cuspidatum against carbon tetrachloride-induced
damage biomarkers during aging in F2 hybrid mice: Protection by liver injury in mice. Plos One [Online]. Available from: http://
chronic oral intake of resveratrol. Free Radical Bio Med 46:799809. www.plosone.org/article/info%3Adoi%2F10.1371%2Fjournal.pone.
Wong VWS, Wong GLH, Chan AWH, et al. (2013). Treatment of non- 0046574 [last accessed 23 Jun 2014].
alcoholic steatohepatitis with Phyllanthus urinaria: A randomized Zhang A, Sun H, Wang X. (2013a). Recent advances in natural products
trial. J Gastroenterol Hepatol 28:5762. from plants for treatmen of liver diseases. Eur J Med Chem 63:5707.
Xavier JR, Gnanam R, Murugan MP, Pappachan A. (2012). Clonal Zhang YX, Hai J, Cao M, et al. (2013b). Silibinin ameliorates steatosis
propagation of Phyllanthus amarus: A hepatoprotector. Pharmacogn and insulin resistance during non-alcoholic fatty liver disease
Mag 8:7882. development partly throughtargeting IRS-1/PI3K/Akt pathway. Int
Xiong SQ, Salazar G, Patrushev N, Alexander RW. (2011). FoxO1 Immunopharmacol 17:71420.
mediates an autofeedback loop regulating SIRT1 expression. J Biol Zhao LT, Wu JQ, Yang JJ, et al. (2011). Dietary quercetin supplemen-
Chem 286:528999. tation increases serum antioxidant capacity and alters hepatic gene
Yasui S, Fujiwara K, Tawada A, et al. (2011). Efficacy of intravenous expression profile in rats. Exp Biol Med 236:7016.
glycyrrhizin in the early stage of acute onset autoimmune hepatitis. Zhou HY, Beevers CS, Huang SL. (2011). The targets of curcumin. Curr
Digest Dis Sci 56:363847. Drug Targets 12:33247.
Downloaded by [202.67.40.194] at 09:32 06 October 2017

Vous aimerez peut-être aussi