You are on page 1of 13

Review Article

Curcumin and Liver Disease Laura Vera-Ramirez1,2


rez-Lopez3
Patricia Pe
Alfonso Varela-Lopez3
MCarmen Ramirez-Tortosa4
Maurizio Battino5
1
GENyO Center Pfizer-University of Granada & Andalusian Government Jose L. Quiles3*
Centre for Genomics & Oncology, Granada, Spain
2
Department of Oncology, Complejo Hospitalario de Jaen, Jaen, Spain
3
 Mataix Verdu
Institute of Nutrition and Food Technology ‘‘Jose  ’’,
Department of Physiology, University of Granada, Granada, Spain
4
 Mataix Verdu
Institute of Nutrition and Food Technology ‘‘Jose  ’’,
Department of Biochemistry and Molecular Biology II,
University of Granada, Granada, Spain
5
Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche,
Facolta di Medicina, Universita Politecnica delle Marche, Ancona, Italy

Abstract
Liver diseases pose a major medical problem worldwide and carcinogenesis and other age-related processes, is its potent
a wide variety of herbs have been studied for the manage- antioxidant activity, which affects multiple processes and
ment of liver-related diseases. In this respect, curcumin has signaling pathways. The effects of curcumin on NF-jb are
long been used in traditional medicine, and in recent years it crucial to our understanding of the potent hepatoprotective
has been the object of increasing research interest. In com- role of this herb-derived micronutrient. Because curcumin is
bating liver diseases, it seems clear that curcumin exerts a a micronutrient that is closely related to cellular redox
hypolipidic effect, which prevents the fatty acid accumulation balance, its properties and activity give rise to a series of
in the hepatocytes that may result from metabolic imbalan- molecular reactions that in every case and biological
ces, and which may cause nonalcoholic steatohepatitis. situation affect the mitochondria. C
V 2013 BioFactors,
Another crucial protective activity of curcumin, not only in 39(1):88–100, 2013
the context of chronic liver diseases but also regarding

Keywords: curcuma; cirrhosis; cytokines; fibrosis; hepatology;


inflammation; mitochondria; nuclear factors; oxidative stress;
steatohepatitis; steatosis

1. Introduction Asia, the main concerns are viral and parasitic infections. More-
over, unlike other major causes of mortality, liver disease rates
Liver diseases represent a major medical problem all over the are increasing rather than declining [1]. The liver is the largest
world. In Europe and North America, alcohol abuse and overnu- internal organ and it performs a great number of functions, sup-
trition are the major causes of pathological conditions of the ported by its different cell types [2,3]. Hepatocytes, present at
liver (with viral hepatitis increasing), whereas in Africa and the hepatic parenchyma, represent around 70% of the liver cell
population. Hepatocytes are implicated in the secretion of pro-
teins and bile, in cholesterol metabolism, and in the metabolism
C 2013 International Union of Biochemistry and Molecular Biology, Inc.
V
Volume 39, Number 1, January/February 2013, Pages 88–100 of glucose and glycogen. Detoxification, the metabolism of urea,
*Address for correspondence to: Jose  L. Quiles, Ph.D., Instituto de acute phase response, and blood clotting are other functions
 n y Tecnologı́a de los Alimentos ‘‘Jose
Nutricio  Mataix Verdu ’’, Universidad performed by hepatocytes. Cholangiocyte or bile duct cells are
de Granada, Centro de Investigacio  n Biome dica, Parque Tecnolo  gico de present in the duct epithelium and represent around 3% of the
Ciencias de la Salud, Lab. 120, Avenida del Conocimiento s/n, 18071 liver cell population. They transport bile, control the rate of bile
Granada, Spain. Tel.:þ34 958241000, Ext. 20316; Fax:þ34 958241000 Ext.
20316; E-mail: jlquiles@ugr.es.
flow, and secrete water and bicarbonate to control the pH of
Received 30 July 2012; accepted 13 September 2012
bile. Endothelial cells are present at the liver vasculature, con-
DOI: 10.1002/biof.1057
trolling blood flow and contributing to parenchymal zonation.
Liver sinusoidal endothelial cells constitute around 2.5% of the
Published online 10 January 2013 in Wiley Online Library
(wileyonlinelibrary.com) lobular parenchyma and form the sinusoidal plexus to facilitate

88 BioFactors
Main liver diseases
TABLE 1

Neoplastic liver Hepatocellular carcinoma


diseases
Hepatoblastoma
Cholangiocarcinoma
Primary hepatic angiosarcoma
Non-neoplastic Alcohol-related disease
liver diseases
Hepatic cholestasis
Nonalcoholic fatty liver diseases (NAFLD)
Nonalcoholic steatohepatitis (NASH)
Human hepatitis viruses

The liver lobule. Hepatic toxicity


FIG 1
Toxicity mediated by drugs
Iron-induced hepatic toxicity
blood circulation. They also enable the transfer of molecules
Thioacetamide-induced hepatic injury
and proteins between serum and hepatocytes, and are active in
the scavenging of macromolecular waste, in cytokine secretion, Carbon tetrachloride toxicity
antigen presentation, and blood clotting. Hepatic stellate cells
Hepatic fibrosis, cirrhosis, and portal
(HSCs) are situated in a perisinusoidal position and represent
hypertension
around 1.4% of liver cells. Their functions are the maintenance of
the extracellular matrix, vitamin A storage, control of vascular
tone, contribution of regenerative response to injury, the
secretion of cytokines, and to act as precursors of myofibroblasts. A large number of molecules have been studied in
The activation of HSCs is the main event for cell-mediated mech- attempts to manage liver-related diseases. Many of these
anisms of liver injury [4]. Kupffer cells, which are present in the drugs are of herbal origin and in some aspects they are still
sinusoids and comprise around 2% of the liver, work by scaveng- not well characterized. Curcumin has long been used in
ing foreign material and secreting cytokines and proteases. traditional medicine and research into this molecule has
Finally, pit cells are natural killer cells present in the liver; these increased considerably in recent years. The main aim of this
are very rare and perform cytotoxic activity. review is to summarize the most important actions of curcu-
To understand liver pathology, it is necessary to briefly con- min in liver diseases, focusing on its mechanism of action with
sider the structure of the adult liver. This organ might appear respect to various pathological conditions. As another chapter
to be a mere pool of hepatocytes arranged within a low level of this book is devoted to curcumin and cancer, we will discuss
histological structure. However, the liver has a very complex here mainly non-neoplastic liver diseases.
architecture organized around its basic unit, the liver lobule
(Fig. 1). The liver lobule [5] is a hexagonal-like structure sur-
rounding the central vein. Hepatocytes are arranged in arrays 2. Liver Diseases
one or two cells thick. Sinusoids are special blood vessels
formed between two arrays lined with sinusoidal cells connect- The basic etiologies of liver injuries are presented in Table 1.
ing the portal venous system to the systemic venous system.
Kupffer cells (liver-resident macrophages) are located in the 2.1. Neoplastic Malignancies
sinusoids, and portal triads are arranged around the lobule. Liver tumors, although not the most frequent, arouse great in-
Triads consist of the termination of the portal vein branches, terest. Research in this field expands continuously, and signifi-
hepatic artery branches, and a bile duct. Between two hepato- cant advances are being achieved in diagnosis and treatment
cytes, small bile ducts constitute the first passage for bile exer- [6]. Hepatocellular carcinoma is one of the most common such
tion by hepatocytes. Stellate cells are located in the space of malignancies and one of the principal complications in patients
Disse, between the hepatocytes and the sinusoidal cells. with chronic liver disease or cirrhosis related to hepatitis B or

Vera-Ramirez et al. 89
BioFactors

C virus infection [7,8]. Hepatoblastoma is a rare childhood ities in hepatocyte transport of bile constituents, due to gene
cancer that presents low rates of incidence and is difficult to alterations in synthesis or transporter mechanisms, such as
study. Nevertheless, different types and subtypes of hepato- progressive familial intrahepatic cholestasis syndrome, ATP8B1
blastoma have been identified [9,10]. Cholangiocarcinoma disease, or ABCB4 disease. Other syndromic forms of inherited
affects the biliary tract epithelium, and it may be present in in- cholestasis include intrahepatic biliary hypoplasia, familial
trahepatic or extrahepatic biliary ducts. Its incidence has hypercholanemia, or lymphedema-cholestasis syndrome [13].
increased in recent years, as have diagnostic mechanisms [11].
Primary hepatic angiosarcoma is a tumor of vascular endothe-
lial cell origin. This represents only 1.8% of all hepatic dis- 2.4. Nonalcoholic Fatty Liver Diseases
eases, and so it has not been as studied as much as other he- Nonalcoholic fatty liver diseases (NAFLD) range from hepatic
patic pathologies [12]. steatosis, the most clinically benign, through nonalcoholic stea-
tohepatitis (NASH), an intermediate lesion, to cirrhosis. Steato-
sis, NASH, fibrosis, and cirrhosis can result from metabolic
2.2. Alcohol-Related Disease
abnormalities and/or genetic predisposition, whereas NAFLD is
Alcoholism continues to affect ever more people worldwide,
associated with obesity, insulin resistance, and type 2 diabetes.
and it is a major cause of mortality in the United States and
Studies have suggested that obesity-related fatty liver disease is
Europe. In disease prediction, practically all cases involve fatty
related to metabolic syndrome [19,20]. NASH, the most widely
liver (known as steatosis), because of the excessive accumula-
studied NAFLD and which was first described as a clinical entity
tion of triglyceride in the hepatocytes. More than one in five
by Ludwig et al. [21], is a precursor to more severe liver dis-
alcoholics develop alcoholic hepatitis, and one-third of these
ease, and in almost 25% of patients it evolves into cirrhosis or
will suffer cirrhosis, which may develop to hepatocellular carci-
even hepatocellular carcinoma. In addition, it represents a fre-
noma, depending on the intensity and duration of alcohol
quent cause of abnormal liver test results in blood donors, and
intake, gender, and genetic and epigenetic factors that affect
is responsible for the asymptomatic elevation of serum amino-
alcohol metabolism and elimination in the liver. Alcoholic hepa-
transferases in over half of cases [22].
titis takes place because alcohol compromises the intestinal
The histological features of NASH have been well described,
barrier, leading to an increased presence of bacteria-derived li-
and in 1999, Brunt et al. [23] proposed a system to classify the
popolysaccharide (LPS) in the portal blood, which produces an
global assessment of necroinflammatory activity (grade) and fi-
excessive and dysregulated inflammatory response, resulting in
brosis with or without architectural remodeling (stage). This
hepatocyte injury and tissue necrosis [13,14]. One of the main
classification included three categories or grades of NASH nec-
alcohol effects on hepatocytes is oxidative stress. Ethanol indu-
roinflammation: grade 1 (mild), grade 2 (moderate), and grade
ces the generation of reactive oxygen substances (ROS) in hepa-
3 (severe), which correlate, respectively, with hepatocellular
tocytes, in the mitochondria, and also in cytosol, where free
steatosis, ballooning, and disarray, together with inflammation
iron is present together with enzymes such as xanthine oxidase
(acinar and portal). According to this classification, fibrosis is
and aldehyde oxidase. Besides ROS production, alcohol prod-
constituted of four categories (stages 1–4), depending on the
ucts can damage hepatocyte antioxidant defense components,
increase in connective tissue deposition and architectural
both enzymatic (such as superoxide dismutase, catalase, or glu-
remodeling. However, the pathogenesis of NASH is still not fully
tathione transferase) and nonenzymatic (principally metal-
understood. The theory currently most accepted is ‘‘the two-hit
binding proteins and vitamins) [13,14].
hypothesis,’’ proposed by Day and James [24]. The ‘‘first hit’’ is
the development of steatosis, following prolonged overnutrition
2.3. Hepatic Cholestasis
that deregulates the lipid metabolism and provokes an accumu-
Hepatic cholestasis refers to the situation of damaged bile
lation of free fatty acids (FFAs) and triglycerides in the liver.
secretion in the liver, characterized by the blockage of bile flow
The increased presence of FFAs in hepatocytes reduces b-oxi-
from the hepatocyte to the intestine, with the consequent accu-
dation, and thus enhances the accumulation of fatty acids. In
mulation of hydrophobic bile acids in the liver and plasma. Its
the ‘‘second hit,’’ steatosis progresses to inflammation and fi-
effects are commonly oxidative stress, increased apoptosis, and
brosis due to oxidative stress, mitochondrial dysfunction, and
fibrosis [15]. Hepatic cholestasis is strongly related to mitochon-
inflammatory cytokines, leading to liver cell inflammation and
drial dysfunction, because the mitochondria mediate death re-
necrosis, and activating the fibrogenic cascades [20,22,25].
ceptor signaling, contributing to oxidative damage and also to
fibrosis, whereas inflammation has been linked with apoptosis
[15]. Hepatic cholestasis is provoked by diverse inducing fac- 2.5. Human Hepatitis Viruses
tors, including drug treatment, bile duct ligation, and inherited There are five human hepatitis viruses, A–E, each with a dif-
and syndromic forms [16,17]. Experiments with rats have ferent development of infection. Hepatitis A and E are always
shown that bile duct ligation perturbs liver homeostasis, raising transient, whereas Hepatitis B, C, and delta may be either
levels of cytokines and signaling molecules; eventually, it may transient or chronic. Despite their differences, they all have
induce cirrhosis [18]. Inherited forms of intrahepatic cholestasis important features in common: their primary target of infec-
include disorders of primary bile acid synthesis and abnormal- tion is the hepatocyte, where they infect and replicate,

90 Curcumin and Liver Disease


although the mechanisms of viral infection and pathogenesis periodic lower doses, accompanied by high levels of ROS, the
vary depending on the hepatitis virus [13]. production of proinflammatory molecules, and the activation
During the acute phase of hepatitis, for 2–6 weeks, many of HSCs [18,30].
hepatocytes are infected and viruses may enter the blood In humans and animals, hepatotoxicity is the major out-
stream or the bile canaliculi. In this acute phase, the immune come of exposure to carbon tetrachloride (CCl4). Liver injury is
system attempts to eliminate the virus, primarily by antibodies detectable by clinical signs (jaundice, swollen, and tender
specifically directed against viral antigens. In a second step, in- liver), biochemical alterations (elevated levels of hepatic
tracellular viruses are eliminated by the cellular immune enzymes in the blood and loss of enzymatic activities in the
response, via the immune destruction of infected cells by cyto- liver), or histological examination (fatty degeneration and ne-
toxic T lymphocytes (CTLs). This second step can also be crosis of central hepatocytes, destruction of intracellular or-
achieved by noncytolytic elimination, by antiviral cytokines that ganelles, fibrosis, and cirrhosis). In the absence of clinical
inhibit viral gene expression and replication, and which there- signs, elevated levels of serum enzymes such as alanine ami-
fore do not need to destroy the infected cell. Chronic liver dis- notransferase (ALT), aspartate aminotransferase (AST), alka-
ease appears when the immune system fails to eliminate the vi- line phosphatase, and gamma glutamyl transferase may pro-
rus infection [13]. Hepatitis B virus (HBV), hepatitis C virus vide evidence of hepatocellular injury [31].
(HCV), and hepatitis delta virus (HDV) are potentially the most
harmful for the liver, being responsible for a large proportion of 2.7. Hepatic Fibrosis, Cirrhosis, and Portal
liver failure conditions, such as fulminant hepatitis, cirrhosis, Hypertension
and hepatocellular carcinoma [26]. HBV infection can be tran- Many of the above pathological conditions can lead to hepatic fi-
sient, in the form of acute hepatitis or fulminant hepatitis, or brosis and cirrhosis or are closely related to portal hyperten-
develop to become chronic, leading to cirrhosis and/or hepato- sion-related events. Fibrosis is characterized by an excessive
cellular carcinoma [27]. Regarding HCV, up to 75% of persons accumulation of extracellular matrix proteins, including colla-
infected progress to chronic hepatitis C. Most of these patients gen. The main collagen-producing cells in the liver are HSCs,
are asymptomatic and present no physical signs of chronic liver which can be stimulated by ROS, inflammatory cytokines, and
disease, other than fatigue. Chronic hepatitis C often progresses apoptotic signals, among others. When this occurs, the hepatic
to cirrhosis and hepatocellular carcinoma [7,13]. architecture is altered; a fibrous scar appears and nodules of
regenerating hepatocytes develop. If the damage continues, the
2.6. Hepatic Toxicity hepatic fibrosis advances to become hepatic cirrhosis, which
More than 1,000 drugs can induce hepatic toxicity, and the risk cannot be reversed. However, recent experimental findings sug-
increases when the daily dose is higher than 50 mg or when the gest that even advanced fibrosis may be reversible
liver is highly involved in metabolizing the drug. The principal [13,17,32,33]. Cirrhosis is the last pathological feature of
mechanism of drug-induced liver damage is that of mitochon- chronic hepatic injury and is characterized by the destruction of
drial dysfunction and lipid dysmetabolism, caused by the drug the hepatic architecture and vascular structures, together with
itself and/or by the reactive metabolites generated. Mitochon- blood flow failure due to the excessive deposition of collagen in
drial dysfunction can have other harmful consequences, such the cirrhotic liver, resulting from an imbalance between colla-
as oxidative stress, energy deficiency, steatosis by accumulation gen synthesis and degradation by membrane-bound metallopro-
of triglycerides, and cell death. In association with obesity and teinase [17,32,33]. Portal hypertension in the liver is the hemo-
diabetes, drugs may induce acute liver injury, steatosis, and dynamic disorder that is most frequently associated with
even steatohepatitis, which can lead to cirrhosis [28]. cirrhosis. Cirrhosis causes distortion of the hepatic architecture,
Another type of hepatic toxicity is iron induced. Iron is an followed by increased intrahepatic resistance and, finally, portal
essential constituent of the body that participates in a large hypertension, which is associated with other clinical pathologies
number of biological reactions. It is present in functional form that may affect diverse organs. Such pathologies include diges-
in molecules such as cytochromes, hemoglobin, myoglobin, tive hemorrhage, hypertensive gastropathy, hepatorenal and
and iron-dependent enzymes, and a good iron balance is nec- hepatopulmonary syndromes, and alterations in the elimination
essary to avoid diseases. Iron overload may be caused by a of drugs, microorganisms, and chemical substances by the liver
wide range of acquired and hereditary conditions, such as he- [13,34]. In addition to changes in liver architecture caused by
reditary hemochromatosis [29], and this condition is associated cirrhosis, the first step to portal hypertension is an increased re-
with various toxic effects, of which the most common is liver sistance to portal blood flow due to deficit and hyporesponse to
damage, with the excessive deposition of iron giving rise to fi- vasodilators, such as nitric oxide (NO), carbon monoxide (CO),
brosis and cirrhosis [18]. or hydrogen sulfide (H2S). The increased production of and
The capacity of the liver to metabolize and help excrete response to vasoconstrictors, such as endothelin, angiotensin II,
xenobiotics makes it very susceptible to damage, which may or alpha-adrenergic stimulus, also promotes hepatic vascular
be reversible but may also lead to fulminant hepatic failure resistance. This imbalance between excessive vasoconstrictors
and death. The administration of high doses of thioacetamide and deficient vasodilators leads to an increase in portal blood
(TAA) to rats causes fibrosis, whereas cirrhosis is provoked by flow that aggravates portal hypertension [13].

Vera-Ramirez et al. 91
BioFactors

3. Mechanisms Involved in Liver Kupffer cells and blood-derived neutrophils, monocytes/macro-


phages, and lymphocytes all release growth factors, cytokines,
Disease chemokines, and ROS, which in turn activate HSCs into becom-
Various in vitro and in vivo observations suggest that oxidative ing MFs [37,40].
stress and associated DNA, protein, and lipid damage may Early MF activation involves the rapid induction of b plate-
underlie liver diseases as a key pathophysiological force and let-derived growth factor (b-PDGF) receptor, the development
which may also be related to chronic liver injury, hepatic of a contractile and fibrogenic phenotype, and the modulation
inflammation, and fibrosis [35]. of growth factor signaling. The autocrine PDGF loop is among
Oxidative stress is a physiological consequence of the dis- the most potent of all cytokine loops during HSC activation.
turbances in the oxidative balance caused by the interplay The union of PDGF with its b receptor activates focal adhesion
between the production of free radicals and the activity of the kinase (FAK), phosphatidylinositol 3-kinase (PI3K), and extrac-
antioxidant defense system. Free radicals are inherent to aero- ellular regulated kinase (ERK) signaling, and hence HSC prolif-
bic life, and are produced and involved in important cellular eration [41]. PDGF signaling is sustained through a positive
processes such as the immune response, vascular biology, thy- feedback mechanism that involves ROS. When the PDGF binds
roid hormone biosynthesis, and oxygen tension sensing. Conse- to its receptor, PI3K and Rac are activated, and this in turn
quently, they are key mediators of physiological cell activity; activates nonphagocytic membrane NADPH-oxidase (NOX) and
however, when the production of free radicals exceeds the ROS production. Among other mitogens that promote HSC pro-
capacity of the antioxidant system to cope with oxidative cellu- liferation and activation through NOX activation, those of vas-
lar and tissue damage, oxidative stress occurs. Diverse conse- cular endothelial growth factor (VEGF), thrombin and its re-
quences arise from persistent oxidative stress, because free ceptor, interleukin (IL)-1, epidermal growth factor (EGF),
radicals alter the chemical structure of any cell component but angiotensin II (Ang II), and basic fibroblast growth factor
they also play a crucial role in cell signaling as second mes- (bFGF) are very significant [42–44]. Other important mitogens
sengers. Thus, an imbalance in the cellular and/or extracellu- for HSCs are tumor necrosis factor a (TNFa) and IL-6, which
lar concentration of free radicals contributes significantly to induce differentiation and activation of the HSCs through the
organ injury and dysfunction, and may ultimately lead to life- activation of a redox-sensitive transcription factor nuclear fac-
threatening degenerative diseases [36]. tor jb (NF-jb), which also plays a prominent role in liver
Among the latter, non-neoplastic liver diseases secondary inflammation, as discussed below [44].
to inflammation and fibrosis are evidently connected with A second event in HSC activation involves migration to-
chronic oxidative stress. Liver fibrosis and its end-stage cirrho- ward injury sites, driven by chemoattractants including PDGF
sis are the consequences of chronic liver injury of varied etiol- and VEGF, which induce the formation of cell protrusions
ogy, including the liver diseases summarized in Section 2. through protein kinase D1 [45], cytoskeletal reorganization
Proinflammatory insults originate from viral, toxic, metabolic, through protein kinase C (PKC) and Rho-dependent signaling
or autoimmune processes and lead to the activation of HSCs, [46], CXCR3 ligands by activation of the Ras/ERK cascade [47],
which transdifferentiate from a quiescent lipid-storing pheno- and monocyte chemoattractant protein 1 (MCP-1) to recruit
type to an a-smooth muscle actin (a-SMA) positive phenotype. and activate monocytes [48]. MCP-1 is mainly involved in the
Activated HSCs are myofibroblast-like cells (MFs) that favor formation and maintenance of the inflammatory infiltrate in
extracellular matrix deposition materials, mainly collagen I, as various wound-healing conditions, including chronic liver dis-
part of the liver’s wound healing mechanism. Portal fibroblasts eases. MCP-1 is overexpressed by a wide range of hepatic cells
and bone marrow-derived mesenchymal stem cells are also including injured hepatocytes [49], activated macrophages and
transdifferentiated into MFs by chronic liver injury. Together, Kupffer cells, activated MFs, and activating HSCs [50] subse-
these cells originate a homogeneous proliferating, contractile, quent to the induction of redox transcription factors such as
and fibrogenic MF cell population regardless of its cellular ori- NF-jb and activation protein 1 (AP-1) [51]. Although HSCs and
gin, in terms of activation and functionality [37]. MFs also con- activated MFs play a significant role in chronic liver diseases
tribute to liver fibrosis by releasing profibrogenic and proin- because of their highly fibrogenic activity, a wide variety of
flammatory cytokines and tissue inhibitors of metalloproteases immune and inflammatory cells are also recruited to injury
[38,39]. This progressive deposition of extracellular matrix sites, forming the inflammatory infiltrate, including mononu-
leads to hepatic structural and functional impairment. clear cells, Kupffer cells, sinusoidal endothelial cells, platelets,
T-lymphocytes, and endothelial progenitor cells [52]. As men-
tioned above, mediators of oxidative stress, such as ROS and
3.1. Oxidative Stress and Inflammation other reactive species, may contribute to injury continuity, by
Among the mechanisms involved in mediating the process of triggering and regulating the expression of proinflammatory
liver fibrosis, an important role is played by those mediated by cytokines and chemokines in MFs and immune cells. The main
ROS, because persisting liver injury leading to parenchymal signaling pathway inducing this activity is mediated by the
damage and hepatocyte loss induces the recruitment of activation of NF-jb, a major redox-sensitive transcription fac-
immune effectors at the injured site. Activated resident tor, which binds to specific DNA regions known as jb

92 Curcumin and Liver Disease


sequences and activates the expression of genes involved in without cytochrome c release, provoking necrotic and caspase-
inflammation, cell survival, proliferation, and differentiation in independent cell death [62] Upon these events, the ROS-medi-
response to different stimuli, including the presence of free ated and sustained activation of c-Jun amino-terminal kinases
radicals [53]. The injury-induced activation of hepatic NF-jb (JNKs), through the oxidative inhibition of JNK phosphatases,
promotes the production and secretion of TNFa, IL-6 in may also reinforce hepatocyte apoptosis [59].
Kupffer cells [52] and the expression of inducible NO synthase Severe oxidative stress secondary to an acute inflamma-
(iNOS) and cycloxygenase-2 in monocytes [54]. Generally, any tory response in liver injury may then provoke a molecular
cytokine capable of activating NF-jb is likely to induce ROS shift that involves the transition from survival signals to death
generation, which further increases the inflammatory signals in injured hepatocytes. This in turn releases further
response. On the other hand, oxidative products such as 4- ROS to the injury site, reinforcing the inflammatory response
hydroxy-2,3-nonenal have been shown to upregulate the and activating HSCs in a paracrine fashion. The result is a
expression of tumor growth factor (TGF-b1) in Kupffer and chronic inflammatory process that contributes to perpetuate
macrophages [55] and to promote leukocyte chemotaxis [56]. fibrogenic progression through sustained MF activation.
Additionally, intrahepatic hypoxia may occur during inflamma-
tory and fibrotic processes, and indeed, the role of proangio- 3.3. Fibrogenesis
genic cytokines and the expression of related receptors in liver Fibrogenesis is the event preceding severe liver malfunction.
fibrosis is a determinant factor in advanced liver cirrhosis Activated MF cells produce and deposit large amounts of colla-
[57]. At this stage, the role of injured hepatocytes, which are gen type I fibers and, to a much lesser but still significant
prone to apoptosis and necrosis through mitochondrial impair- extent, collagen type III at the liver injury site. Fibrogenic ac-
ment and ROS release, is of crucial importance for injury per- tivity is mainly mediated by TGFb-1, which typically acts by
petuation and liver disease progression to cirrhosis. binding TGF-b type II receptors. Subsequent TGF-b type I acti-
vation and Smad protein association and phosphorylation
3.2. Apoptosis result in a heterodimeric complex that translocates to the nu-
As mentioned above, with parenchymal damage and inflam- cleus and binds to specific nucleotide motifs at the promoter
matory signaling, the injury focus is a complex and highly region of the collagen, type I, alpha 1 (COL1A1) gene [63].
oxidative environment composed of inflammatory and angio- Additionally, TGFb-1 induces the accumulation of hydrogen
genic cells releasing a wide variety of growth factors and peroxide (H2O2), which is involved in upregulating the expres-
inflammatory cytokines and chemokines. Among the latter, sion of the COL1A1 gene through a CCAAT/enhancer binding
TNFa activates molecular pathways, which leads to either pro- protein-b (C/EBPb)-dependent mechanism in MFs [64]. The
survival or to proapoptotic and necrotic signals, depending on TGFb-1 fibrogenic signal is also mediated by connective tissue
the redox state of the hepatocyte. growth factor (CTGF), which is normally expressed by MFs,
The interaction of TNFa with its type 1 receptor (TNFR1) is endothelial cells, and ductular epithelial cells but also by
followed by the association of the TNF-receptor-associated parenchymal hepatocytes [65]. Moreover, CTGF expression in
death domain (TRADD), the Rieske iron-sulfur protein of parenchymal hepatocytes appears to be both TGFb-1-depend-
mitochondrial ubiquinol-cytochrome c reductase (RIP1), and ent and TGFb-1-independent, and this process can also be up-
TNF-receptor-associated factors 2 and 5 (TRAF-2 and TRAF-5). regulated by a wide variety of factors, including lipid peroxida-
These interactions activate NF-jB and AP-1 transcription fac- tion products (hydroxynonenal and malondialdehyde) [66].
tors, which in turn activate the expression of genes that pro- Finally, the contraction of active fibrogenic MFs may con-
mote hepatocyte survival [58]. However, glutathione (GSH) tribute to intrahepatic resistance, portal hypertension, and lob-
depletion alters the susceptibility of hepatocytes to TNF-a and ular distortion as fibrosis advances and cirrhosis develops. MF
promotes the TNF-a-induced apoptosis axis, which finally leads contraction activity is mediated by both Caþ2-dependent mech-
to mitochondrial dysfunction, promoting the overproduction of anisms such as the myosin light chain kinase (MLCK) pathway,
ROS, impaired bioenergetics, severe oxidative stress, and cell and by Caþ2-sensitization mechanisms such as 17-kDa PKC-
death [59,60]. Under these circumstances, the TRADD/RIP-1/ potentiated protein phosphatase 1 inhibitor protein (CPI-17)
TRAF-2 complex can dissociate from TNRF1 and bind Fas- and myosin light chain phosphatase targeting subunit 1
ligand-associated death domain (FADD), recruiting caspase 8/ (MYPT1) pathways [67].
10, which cleaves the proapoptotic protein BH3 interacting do- From this summary of the most active and important sig-
main death agonist (Bid). The cleaved tBid form translocates to naling pathways that intervene in liver fibrosis, it is possible to
the mitochondria, producing the permeabilization of the mito- envisage a highly oxidative environment in which an inducible
chondrial outer membrane, the release of cytochrome c, and and complex cell population receives a wide range of signals
the activation of the classic or intrinsic apoptosis pathway and from the extracellular milieu, such as growth factors, cyto-
ROS production [41,61]. kines, and ROS or other reactive species, which promote
Alternatively, upon TNFa stimulation, RIP1 may be capable inflammatory and fibrogenic processes. In such a highly induc-
of translocating to the mitochondria and permeabilizing the ible and oxidative microenvironment, immune and angiogenic
mitochondrial outer membrane, thus eliciting ROS release, cells unavoidably increase their intracellular concentration of

Vera-Ramirez et al. 93
BioFactors

ROS, which significantly affects cellular signaling pathways, tein-2 (UCP-2) is thought to activate increasing proton leaks
increasing disease severity and finally leading to liver degener- across the mitochondrial inner membrane, reducing mito-
ation and cirrhosis. In this sense, it is well known that protein chondrial inner transmembrane potential (DWm) and oxida-
tyrosine phosphatases (PTPs) are susceptible to oxidation tive phosphorylation. This protective effect against ROS pro-
because of chemical changes in the conserved cysteine residue duction may in turn favor further triglyceride accumulation
of their catalytic domain that possesses a low pKa. This reac- in the hepatocytes. This topic remains controversial in mito-
tion inhibits phosphatase activity, and thus increases cytoplas- chondrial bioenergetics, and further data are required to
mic protein kinase activity and stimulates the wide range of in- clarify the question [74–76].
tracellular signaling cascades that are mediated. Among these Many of these changes are mediated by the activation of
molecular pathways, those initiated by inflammatory cytokines nuclear peroxisome proliferator-activated receptor-c (PPAR-c),
and growth factors can be counted [68,69]. These important which promotes the expression of enzymes involved in lipid ca-
changes affect MF and inflammatory cells, as well as hepato- tabolism in the mitochondria and in the peroxisomes. Interest-
cytes, increasing profibrotic signaling and reinforcing ROS- ingly, PPAR-c is activated by long-chain FFA, which may be
mediated signaling in a cyclic fashion. With respect to the lat- the cause of the observed rise in the expression of this tran-
ter, mitochondrial damage plays a pivotal role, acting as an scription factor in obesity and steatosis models [77,78].
‘‘amplifying effecter’’ in a key stage of liver disease, and mak- Thus, defects in lipid homeostasis, bioenergetic impair-
ing a major contribution to the transition from acute injury to ment, and the overproduction of ROS, leading to lipid accumu-
chronic damage through hepatocyte cell death and ROS lation and oxidative stress due to severe hyperlipemia, all
release. induce massive hepatocyte apoptosis, which provokes an acute
inflammatory response, progressing to liver necrosis,
3.4. Hyperlipemia increased and chronic inflammation, and liver fibrosis. NASH
As stated above, a high level of circulating and hepatic FFAs and NAFLD are classic examples of chronic liver diseases
is a common cause of liver steatosis and steatohepatitis. Met- induced by hyperlipemia.
abolic disturbances leading to steatosis (as associated with
obese or overweight patients and with metabolic syndrome,
and often including diabetes, insulin, and leptin resistance) 4. Curcumin in Liver Diseases
lead to a long-term accumulation of triglyceride in the hepa-
tocytes, coupled to ROS production, due to mitochondrial Curcumin [1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-
impairment. This process is described in the ‘‘two hit’’ model 3,5-dione] is a natural yellow polyphenol extracted from the
of NASH published by Day and James, which continues to be rhizome of turmeric (Curcuma longa), a plant grown in tropi-
widely accepted [24]. Mitochondrial dysfunction leads to cal and subtropical regions throughout the world, which is
altered bioenergetics through the impairment of mitochon- extensively used for food preparation in many Asian countries.
drial b-oxidation and FFA accumulation, which in turn induce Besides its culinary use, curcumin has been considered a me-
microsomal FFA oxidation (x-oxidation) involving cytochrome dicinal herb in the traditional medicine of several countries for
P450 isoforms CYP2E1 and CYP4A to compensate for FFA centuries, because of to its antioxidant, anti-inflammatory,
overload. CYP2E1 and CYP4A activity induces the production antimutagenic, antimicrobial, and anticancer properties. How-
of ROS, increasing intracellular oxidative stress and leading ever, the molecular network that governs curcumin-derived
to lipid peroxidation [70]. It has also been shown that ROS effects on health has not been completely elucidated [79].
generated by CYP2E1 activity promotes insulin resistance, Numerous in vitro studies have indicated that curcumin
decreases insulin signaling in the liver, and increases hepatic exerts potent antioxidant and anti-inflammatory properties,
fat accumulation [71]. which may account for its protective effect in chronic liver dis-
The mitochondrial b-oxidation of FFA continues in paral- eases. Importantly, several in vivo studies from animal models
lel and it is not inhibited until massive electron flux on the of chronic liver diseases have shown that curcumin possesses
electron transport chain and ROS overproduction provoke antifibrogenic properties, mediated by the inactivation of dif-
mitochondrial dysfunction. It has been shown that ferent processes involved in liver damage [80].
mitochondrial lipotoxicity is not an early event due to hepatic Early experiments published by Matsuda et al. [81,82]
hyperlipidemia; indeed, initial transient increased mitochon- showed the hepatoprotective role of the main sesquiterpenes
drial activity has been reported in mice fed with hyperlipidic isolated from the aqueous acetone extract of Zedoariae Rhi-
diets; this is then followed by a significant decrease in oxida- zoma, the rhizome of Curcuma zedoaria, in an acute liver
tive phosphorylation due to mitochondrial impairment injury mouse model induced by D-galactosamine (D-GalN)/LPS
[72,73]. As mitochondrial oxidant capacity falls and ROS pro- toxicity. In vitro and in vivo experiments suggested that curcu-
duction increases, lipids accumulate and lipid peroxidation min may play a major role in hepatocytes protection by inhibi-
takes place, further increasing oxidative stress and cellular ting the production of NO and TNF-a in LPS-activated Kupffer
damage. Under these highly stressing circumstances, mito- cells. Later studies using LPS-induced hepatotoxicity further
chondrial uncoupling seems to take place. Uncoupling pro- sustained these findings, highlighting the interfering role of

94 Curcumin and Liver Disease


curcumin on the inflammatory and apoptotic TNF-a/NF-jB sig- the protective activity of curcumin and confirmed the definitive
naling pathway. Lukita-Atmadja et al. [83] showed that pre- involvement of its effects on NF-jB signaling, which remain
treatment with curcuminoids of endotoxemic BALB/C mice sig- paramount to our understanding of the potent hepatoprotec-
nificantly reduced the phagocytic activity of Kupffer cells and tive role of this herb-derived micronutrient.
suppressed the hepatic microvascular inflammatory response Ramirez-Tortosa et al. [22] showed that curcumin supple-
to LPS. On the basis of their histopathologic observations and mentation to experimental rabbits with high-fat-induced NASH
previous experimental data, the authors hypothesized that cur- lowers the grade of NASH and aminotransferase activity, and
cumin anti-inflammatory effects may be mediated by the inhi- raises levels of mitochondrial antioxidants with respect to
bition of the nuclear translocation of NF-jB and its dependent healthy control animals. In addition to these changes, curcu-
proinflammatory cytokines, what may contribute to the signifi- min increased levels of mitochondrial antioxidants, reduced
cant decrease in neutrophils recruitment to portal and central mitochondrial reactive oxygen species, improved mitochon-
venules as well as in the sinusoids of mice treated with curcu- drial function, and lowered levels of TNF-a protein levels, thus
min with respect to control mice. Later on, Kaur et al. [84] inhibiting the key apoptotic TNF-a-NF-jB axis in hepatocytes
and Yun et al. [85] provided further insights into the molecular during initial phases of the inflammatory infiltrate formation
mechanisms of curcumin-derived protection against LPS- and followed by massive HSC activation and fibrosis. Curcumin
LPS/GalN-induced liver injury, respectively, confirming the treatment has also been shown to decrease hepatic NF-jB lev-
involvement of TNF-a inhibition in these processes. Both works els, markers of hepatic inflammation, and hepatomegaly in di-
reported that curcumin administration before induction with abetic and obese mouse models [89]. A significant inhibition of
GalN/LPS or LPS alone prevented the elevation in serum TNF-a NF-jB activity is also the main mechanism by which curcumin
and aminotransferases, reduced hepatic necrosis, and inflam- has been shown to preserve liver tissue integrity in early
mation. Additionally, Kaur et al. confirmed the antioxidant stages of hepatic fibrosis in alcoholic liver injury rats [90]. In a
effect of curcumin by reporting decreased levels of lipid perox- rat model of TAA-induced hepatotoxicity, curcumin has dem-
idation measured as thiobarbituric acid reactive substances onstrated to improve the survival rates of the animals by sig-
(TBARS) and increased GSH and SOD levels in the liver ho- nificantly inhibiting the nuclear binding of NF-jB and the iNOS
mogenates from LPS-challenged rats supplemented with cur- protein expression [91]. Novel formulations of curcumin, as
cumin. In 2011, Cerný et al. [86] contributed with a work NanoCurcTM, showing enhanced bioavailability in liver tissue,
showing that the ameliorative effects of curcumin on liver have rendered lower mRNA levels of TNF-a and IL-6 and
injury in a rat LPS/dGalN model were also mediated by the enhanced antioxidant capacity in liver tissue from mice
positive regulation of antioxidant and cytoprotective enzymes, injected with CCl4 intraperitoneally [92]. Curcumin supplemen-
such as heme oxygenase-1 (HO-1). On the other hand, GalN/ tation of mice fed with the methionine- and choline-deficient
LPS treatment dramatically increased lipid peroxidation levels, diet, which develop steatohepatitis, also showed decreased NF-
NO production, and the expression of NO synthase 2 (NOS-2) jB activation and induction of downstream inflammatory cyto-
in the serum and liver of experimental animals. Curcumin- kines [93]. In vivo experiments with hamsters infected with
derived hepatoprotection was evidenced through the decrease the trematode Opisthorchis viverrini, a liver fluke, and treated
in serum aminotransferases and the elevation of the expres- with praziquantel have showed the induction of several inflam-
sion and activity of HO and the acute drop in mRNA levels of matory cytokines, namely NF-jB, and downstream effectors
NOS-2, NO, both in plasma and liver, and liver CO, which is a such as TNF-a, IL-1b, iNOS, and COX-2, and enhanced levels
product of lipid peroxidation. Increases in plasma bilirubin of oxidative stress, which are significantly ameliorated after
normally reflect the severity of hepatic injury upon hepatotoxic treatment with curcumin [94]. Experimental liver steatosis
GalN/LPS treatment, but, in this work, the authors argue that induced by TNF-a injection in mice showed significant attenua-
further increases in the experimental group supplemented tion after curcumin treatment reflected in decreased oxidative
with curcumin beyond those observed in rats not supple- stress, neutrophils infiltration, and improved histopathology
mented with curcumin are the consequence of HO-1 activity. [95]. Taking into account the heterogeneous sources of liver
Interestingly, bilirubin is a potent antioxidant preventing lipid damage in the models employed by these studies and their in-
peroxidation (which indeed demonstrated to be reduced upon variable confluence on the molecular pathways affected by
curcumin treatment). On the other hand, previous in vitro curcumin in the reduction of damage, it should be pertinent to
studies using RAW264.7 macrophages have suggested that conclude that the inhibition of the NF-jB plays a major role
HO-1 upregulation and the decrease in NO production are regarding the hepatoprotective activity of curcumin, yet its
mediated by the inhibitory effect of curcumin on NF-jB [87], specific molecular mechanisms remain to be elucidated.
and the use of monocarbonyl analogs of curcumin has also As mentioned before, besides TNF-a signaling, curcumin
showed to decrease LPS-induced expression of TNF-a and IL-6 has been reported to inhibit the expression of other NF-jB-
in mouse J774A.1 macrophages [88]. dependent inflammatory chemokines and cytokines, such as
Apart from those works using LPS/dGalN-induced hepatitis IL-6, IL-2, TGF-b, and MCP-1, and inflammation-promoting
models, many other in vitro and in vivo experiments, per- enzymes such as COX-2 and iNOS in Kupffer cells and hepatic
formed in a wide variety of liver injury models, have evidenced tissue homogenates [92,94,96,97]. Particularly, TGF-b is of

Vera-Ramirez et al. 95
BioFactors

special interest during the latest phase of liver injury given its frequently observed in both experimental alcoholic liver dis-
involvement in liver fibrosis. In vivo studies using a bile duct li- ease and in CCl4-induced injury [108,109]. Increased levels of
gation and CCl4 rat models have shown that curcumin is able catalase, glutathione peroxidase, and GSH are also restored
to partially inhibit the expression of TGF-b, significantly pre- after curcumin supplementation in hepatocellular toxicity
venting bile duct ligation fibrosis [98]. Using the rat HSC-T6 models induced by cadmium [109,110] or paracetamol
cell line, Lin et al. [99] showed that curcumin suppressed the [111,112].
expression of SMA and collagen deposition in the HSC-T6 cells Curcumin has been shown to decrease lipid storage in the
after stimulation with TGF-b, accounting for its antifibrogenic hepatocytes of high-fat diet rodents, contributing to minimize
effects. Nevertheless, increased curcumin concentrations eli- the liver damage induced by mitochondrial dysfunction and
cited a different effect, as they induced HSC-T6 cells apoptosis. oxidative stress. Initial data regarding the hypolipemic activity
Then, it has been suggested that curcumin exerts its hepato- of curcumin were reported by Rao et al. [113], who showed
protective effects at this level through different and dose-de- that rats fed concurrently with cholesterol and curcumin pre-
pendent mechanisms. This observation was further corrobo- sented a lower cholesterol content in the liver than did rats
rated in vivo, through a study published shortly after by Shu fed with cholesterol only. Later, Asai and Miyazawa [114]
et al. [100] using a CCl4-induced liver injury rat model. Later, showed that male Sprague-Dawley rats supplemented with 1.0
Nakayama et al. [101] studied the molecular processes govern- g curcuminoids/100 g diet registered significantly lower liver
ing TGF-b-mediated liver fibrosis and curcumin injury preven- triacylglycerol and cholesterol concentrations and significantly
tion using human Hep2G cells. This approach showed that higher hepatic acyl-CoA oxidase activity than did control rats
TGF-b induces the expression of plasminogen activator inhibi- not given any curcumin supplement. This would account for
tor type-1 (PAI-1), a highly profibrotic molecule, through multi- curcuminoids’ lipid-lowering potency in vivo. Rukkumani et al.
ple mechanisms involving NF-jB signaling and oxidative [115,116] assessed the hepatoprotective and hypolipemic
stress. Interestingly, the authors argued that curcumin signifi- effects of curcumin and its synthetic analogs in a male rat
cantly reduced PAI-1 expression, providing a mechanistic ex- model of alcohol-induced and high-fat diet liver injury. In both
planation for the hepatoprotective effects of curcumin regard- experiments, curcumin was shown to attenuate histopathologi-
ing liver fibrosis. Additionally, curcumin has been related to a cal changes in the liver and to reduce hepatic tissue levels of
significant reduction in liver fibrosis and injury induced by cholesterol, triglycerides, and FFAs. More recently, Jang et al.
both CCl4 and Concavalin A through the negative modulation [117] showed that curcumin supplementation in a high-fat diet
of the expression of Toll-like receptor (TLR) 2, TLR4, and in hamsters lowered levels of circulating lipids and hepatic lip-
TLR9, providing further insights into the molecular processes ids. Significantly, they also showed that curcumin increased
involved in liver pathogenesis [102,103]. fatty acid b-oxidation activity and reduced the activity of lipid
Curcumin’s activity, which effectively modulates important anabolic enzymes such as fatty acid synthase, 3-hydroxy-3-
molecular pathways for cell biology and homeostasis, shows the methylglutaryl CoA reductase, and acyl CoA cholesterol acyl-
therapeutic potential of this widely appreciated micronutrient, transferase. The latter property was also highlighted very
not only in the context of chronic liver diseases but also regard- recently by Shao et al. as a molecular mechanism of curcumin
ing carcinogenesis and other age-related processes. Beyond to hepatoprotection [118]. Consequently, decreased levels of he-
its known attenuating role in inflammation and fibrosis signal- patic lipid peroxides were detected in hamsters given a diet
ing, curcumin is well known to exert a potent antioxidant activ- supplemented with curcumin, with respect to control ham-
ity. Indeed, many of the works discussed above include the anal- sters. Finally, curcumin has also been shown to significantly
ysis of several oxidative stress and antioxidant markers to inhibit the expression and activity of key proteins involved in
further elucidate the molecular mechanisms by which curcumin metabolism, such as hepatic protein tyrosine phosphatase 1B
elicits its hepatoprotective activity. (PTP1B), which is associated with defective insulin and leptin
Curcumin has been shown to improve both acute and sub- signaling in a model of hypertriglyceridemia and hepatic stea-
acute liver injury induced by CCl4 intraperitoneal injection in tosis in fructose-fed rats [119], and leptin-like oxidized low-
rats by lowering the activity of serum aminotransferases and density lipoprotein (LDL) receptor-1 (LOX-1) as a result of the
the levels of liver lipid peroxidation [104], reactivating the disruption of Wnt signaling and the activation of PPAR-c.
impaired activity of important antioxidant enzymes, such as Reducing the expression of LOX-1, curcumin has been shown
hepatic SOD, lowering liver lipids and lipid peroxidation, and to block the transport of extracellular ox-LDL into HSCs, thus
increasing total GSH levels [105] Similarly, Pari et al. showed contributing to preventing their activation [120]. It seems clear
that curcumin and tetrahydrocurcumin, one of the main prod- that curcumin exerts a hypolipidic effect, which prevents fatty
ucts of curcumin catabolism, administered before chemical acid accumulation in the hepatocytes as a result of various
induction of hepatocellular damage with erythromycin estolate metabolic imbalances that finally lead to NASH.
or chloroquine effectively reduce serum and hepatic levels of Finally, very recent studies have reported that curcumin
lipid peroxides and TBARS together with a significant increase also inhibits CYP2E1 activity [121], thus limiting ROS produc-
in nonenzymatic and enzymatic antioxidants [106,107]. Impor- tion from microsomes, and activates NF-E2-related factor 2
tantly, curcumin is known to inhibit GSH depletion, which is (Nrf2) translocation to the nucleus, where it activates the

96 Curcumin and Liver Disease


expression of antioxidant enzymes [94,122], elevating the esses are extremely scarce, with many of them involving
expression of the apurinic/apyrimidinic endonuclease1/redox severe side effects [137]. On the other hand, despite the proven
factor-1 (APE1/Ref-1) DNA repair protein [97], and preventing enormous therapeutic potential of curcumin and its safety for
the activation of HSCs through PPAR-c overexpression, which human use, it has not yet been approved for use in the treat-
maintains lipid storing (quiescent) phenotype in these cells and ment of any human disease. Therefore, future research into
blocks PDGF and EGF signaling [123]. It also increases the mitochondrial dysfunction-associated diseases, such as chronic
expression of mitochondrial biogenesis genes such as nuclear liver disease, should focus on large, well-controlled human
respiratory factor 1 (NRF1) and mitochondrial transcription studies to fully develop the therapeutic potential of this
factor A (Tfam) [124]. micronutrient.
Regarding clinical studies assessing the hepatoprotective
activity of curcumin, there are limited data mainly related to
the study of its pharmacokinetics and toxicity [125]. Curcumin Acknowledgements
is poorly absorbed following oral administration, as it is not L. Vera-Ramirez was supported by a postdoctoral contract
hydrosoluble, in such a way that the majority of the ingested from the Andalusian Regional government. P. Perez-Lopez was
curcumin is excreted intact after passing through the digestive supported by a predoctoral FPI grant from the Spanish Minis-
tract. The rest is metabolized at the intestine mucosa and liver pez was supported
try of Science and Innovation. A. Varela-Lo
rendering detectable curcumin plasma levels (0.41–1.75 lM af- by a predoctoral FPU grant from the Spanish Ministry of Sci-
ter 1 h of oral dosing) when administered 4–8 g [126–128]. It ence and Innovation. The authors acknowledge the University
has been shown that curcumin absorption is greatly improved of Granada and the Andalusian Regional government for sup-
by the concurrent oral administration of piperine from black porting research of the group.
pepper [128]. In light of these observations, currently great
efforts are being made to improve curcumin bioavailability
and potentially study and exploit its therapeutic properties. References
This way novel curcumin formulations and complexes are [1] Williams, R. (2006) Global challenges in liver disease. Hepatology 44,
being tested and include monocarbonyl analogs of curcumin 521–526.
[87], polymeric nanoparticle formulations of curcumin [2] Blouin, A., Bolender, R. P., and Weibel, E. R. (1977) Distribution of organelles
and membranes between hepatocytes and nonhepatocytes in the rat liver
[92,129], and curcumin and phospholipids complexes [130]. On
parenchyma. A stereological study. J. Cell. Biol. 72, 441–455.
the other hand, curcumin administration has been shown to [3] Si-Tayeb, K., Lemaigre, F. P., and Duncan, S. A. (2010) Organogenesis and
be fairly safe for humans even at high doses of 12 g/day [131]. development of the liver. Dev. Cell 18, 175–189.
Nevertheless, a recent study has pointed out the potential toxic [4] Friedman, S. L. (2008) Hepatic stellate cells: protean, multifunctional, and
activity of several components of turmeric accompanying cur- enigmatic cells of the liver. Physiol. Rev. 88, 125–172.
[5] Vosough, M., Moslem, M., Pournasr, B., and Baharvand, H. (2011) Cell-based
cumin from an in silico analysis [132], what deserves further
therapeutics for liver disorders. Br. Med. Bull. 100, 157–172.
in vivo studies. [6] Irshad, A., Anis, M., and Ackerman, S. J. (2012) Current role of ultrasound in
chronic liver disease: surveillance, diagnosis and management of hepatic
neoplasms. Curr. Probl. Diagn. Radiol. 41, 43–51.
[7] Cauchy, F., Fuks, D., and Belghiti, J. (2012) HCC: current surgical treatment
5. Conclusions concepts. Langenbecks Arch. Surg. 397, 681–695.
[8] El-Serag, H. B. (2012) Epidemiology of viral hepatitis and hepatocellular car-
Curcumin is a highly pleiotropic and multiactivity molecule cinoma. Gastroenterology 142, 1264–1273.
that is capable of significantly affecting a wide variety of proc- [9] Spector, L. G. and Birch, J. (2012) The epidemiology of hepatoblastoma.
esses, ranging from the catalytic activity of antioxidant and Pediatr. Blood Cancer 59, 776–779.
proinflammatory enzymes to the expression of multiple genes [10] Zimmermann, A. (2005) The emerging family of hepatoblastoma tumours:
from ontogenesis to oncogenesis. Eur. J. Cancer 41, 1503–1514.
related to inflammation and redox biology. Curcumin is a
[11] Anderesen, J. B. and Thorgeirsson, S. S. (2012) Genetic profiling of intrahe-
micronutrient that is closely related to cellular redox balance patic cholangiocarcinoma. Curr. Opin. Gastroenterol. 28, 266–272.
and thus to mitochondrial biology, making it the principal cell [12] Duan, X. F. and Li, Q. (2012) Primary hepatic angiosarcoma: a retrospective
organelle for cellular redox control. Not surprisingly, the prop- analysis of 6 cases. J. Dig. Dis. 13, 381–385.
erties and activity of curcumin give rise to a network of molec- [13] Arias, I. M., Alter, H. J., Boyer, J. L., Cohen, D. E., Fausto, N., et al. (2009)
The Liver Biology and Pathobiology, 5th edn. Wiley-Blackwell, West Sus-
ular reactions that in every case and biological situation affect
sex, UK.
the mitochondria. Fat accumulation is a key process in many [14] Zhu, H., Jia, Z., Misra, H., and Li, Y. R. (2012) Oxidative stress and redox
liver diseases, and various studies have shown that mitochon- signaling mechanisms of alcoholic liver disease: updated experimental and
drial-related oxidative stress can be attenuated by specific die- clinical evidence. J. Dig. Dis. 13, 133–142.
tary fat types, which in addition reduce blood lipids [133–136]. [15] Arduini, A., Serviddio, G., Tormos, A. M., Monsalve, M., and Sastre, J.
(2012) Mitochondrial dysfunction in cholestatic liver diseases. Front. Biosci.
Therefore, the combined use of these dietary fats and curcu-
(Elite Ed) 4, 2233–2252.
min would be an interesting approach to the treatment of liver [16] Longerich, T., Flechtenmacher, C., and Schirmacher, P. (2008) Quality and
pathologies. Specific targeted therapies and/or agents for mito- quantity in hepatopathology. Diagnostic and clinically relevant grading for
chondrial disorders and related diseases or degenerative proc- non-tumourous liver diseases. Pathologe 29, 15–26.

Vera-Ramirez et al. 97
BioFactors

[17] Gieling, R. G., Burt, A. D., and Mann, D. A. (2008) Fibrosis and cirrhosis re- [42] Paik, Y. H., Iwaisako, K., Seki, E., Inokuchi, S., Schnabl, B., et al. (2011) The
versibility-molecular mechanisms. Clin. Liver Dis. 12, 915–937. nicotinamide adenine dinucleotide phosphate oxidase (NOX) homologues
[18] Rivera-Espinoza, Y. and Muriel, P. (2009) Pharmacological actions of curcu- NOX1 and NOX2/gp91(phox) mediate hepatic fibrosis in mice. Hepatology
min in liver diseases or damage. Liver Int. 29, 1457–1466. 53, 173017–173041.
[19] Nagarajan, P., Mahesh Kumar, M. J., Venkatesan, R., Majundar, S. S., and [43] De Minicis, S. and Brenner, D. A. (2007) NOX in liver fibrosis. Arch. Bio-
Juyal, R. C. (2012) Genetically modified mouse models for the study of non- chem. Biophys. 462, 266–272.
alcoholic fatty liver disease. World J. Gastroenterol. 18, 1141–1153. [44] Friedman, S. L. (2008) Mechanisms of hepatic fibrogenesis. Gastroenterol-
[20] Siebler, J. and Galle, P. R. (2006) Treatment of nonalcoholic fatty liver dis- ogy 134, 1655–1669.
ease. World J. Gastroenterol. 12, 2161–2167. [45] Van Lint, J., Rykx, A., Maeda, Y., Vantus, T., Sturany, S., et al. (2002) Protein
[21] Ludwig, J., Viggiano, T. R., McGill, D. B., and Oh, B. J. (1980) Nonalcoholic kinase D, an intracellular traffic regulator on the move. Trends Cell Biol. 12,
steatohepatitis. Mayo Clinic experiences with a hitherto unnamed disease. 193–200.
Mayo Clin. Proc. 55, 434–438. [46] Di Sario, A., Bendia, E., Svegliati-Baroni, G., Marzioni, M., Ridolfi, F., et al.
[22] Ramirez-Tortosa, M. C., Ramirez-Tortosa, C. L., Mesa, M. D., Granados, S., (2002) Rearrangement of the cytoskeletal network induced by platelet-
Gil, A., et al (2009) Curcumin ameliorates rabbit’s steatohepatitis via respi- derived growth factor in rat hepatic stellate cells, role of different intracellu-
ratory chain, oxidative stress, and TNF-a. Free Radic. Biol. Med. 47, lar signalling pathways. J. Hepatol. 36, 179–190.
924–931. [47] Bonacchi, A., Romagnani, P., Romanelli, R. G., Efsen, E., Annunziato, F.,
[23] Brunt, E. M., Janney, C. G., Di Bisceglie, A. M., Neuschwander-Tetri, B. A., et al. (2001) Signal transduction by the chemokine receptor CXCR3, activa-
and Bacon, B. R. (1999) Nonalcoholic steatohepatitis: a proposal for grading tion of Ras/ERK, Src, and phosphatidylinositol 3-kinase/Akt controls cell
and staging the histological lesions. Am. J. Gastroenterol. 94, 2467–2474. migration and proliferation in human vascular pericytes. J. Biol. Chem. 276,
[24] Day, C. P. and James, O. (1998) Steatohepatitis: a tale of two ‘‘hits’’? 9945–9954.
Gastroenterology 114, 842–845. [48] Heymann, F., Trautwein, C., and Tacke, F. (2009) Monocytes and macro-
[25] Polyzos, S. A., Kountouras, J., Zavos, C., and Deretzi, G. (2012) Nonalco- phages as cellular targets in liver fibrosis. Inflamm. Allergy Drug Targets 8,
holic fatty liver disease: multimodal treatment options for a pathogeneti- 307–318.
cally multiple-hit disease. J. Clin. Gastroenterol. 46, 272–284. [49] Ramm, G. A., Shepherd, R. W., Hoskins, A. C., Greco, S. A., Ney, A. D.,
[26] Yoshiyuki, U. (2012) The current endeavors to understand the pathogenesis et al. (2009) Fibrogenesis in pediatric cholestatic liver disease, role of tauro-
of intractable liver diseases. Tohoku J. Exp. Med. 226, 171–175. cholate and hepatocyte-derived monocyte chemotaxis protein-1 in hepatic
[27] Hoofnagle, J. H. and di Bisceglie, A. M. (1997) The treatment of chronic viral stellate cell recruitment. Hepatology 49, 533–544.
hepatitis. N. Engl. J. Med. 336, 347–356. [50] Seki, E., de Minicis, S., Inokuchi, S., Taura, K., Miyai, K., et al. (2009) CCR2
[28] Begriche, K., Massart, J., Robin, M. A., Borgne-Sanchez, A., and Fromenty, promotes hepatic fibrosis in mice. Hepatology 50, 185–197.
B. (2011) Drug-induced toxicity on mitochondria and lipid metabolism: [51] Roebuck, K. A., Carpenter, L. R., Lakshminarayanan, V., Page, S. M., Moy, J.
mechanistic diversity and deleterious consequences for the liver. J. Hepatol. N., et al. (1999) Stimulus-specific regulation of chemokine expression
54, 773–794. involves differential activation of the redox-responsive transcription factors
[29] Siddique, A. and Kowdley, K. V. (2012) Review article: the iron overload AP-1 and NF-kappaB. J. Leukoc. Biol. 65, 291–298.
syndromes. Aliment. Pharmacol. Ther. 35, 876–893. [52] Dienes, H. P. and Drebber, U. (2010) Pathology of immune-mediated liver
[30] Abul, H., Mathew, T. C., Dashti, H. M., and Al-Bader, A. (2002) Level of injury. Dig. Dis. 28, 57–62.
superoxide dismutase, glutathione peroxidase and uric acid in thioaceta- [53] Elsharkawy, A. M. and Mann, D. A. (2007) Nuclear factor-kappaB and the
mide-induced cirrhotic rats. Anat. Histol. Embryol. 31, 66–71. hepatic inflammation-fibrosis-cancer axis. Hepatology 46, 590–597.
[31] ATSDR (2005). Toxicological profile for carbon tetrachloride (update), [54] Park, H. J. and Youn, H. S. Mercury induces the expression of cyclo-
Agency for Toxic Substances and Disease Registry, Atlanta, p. 309. oxygenase-2 and inducible nitric oxide synthase. Toxicol. Ind. Health, in
[32] Schuppan, D. and Afdhal, N. H. (2008) Liver cirrhosis. Lancet 371, 838–851. press. doi: 10.1177/0748233711427048.
[33] Vinken, M. (2012) Gap junctions and non-neoplastic liver disease. [55] Chiarpotto, E., Castello, L., Leonarduzzi, G., Biasi, F., and Poli, G. (2005)
J. Hepatol. 57, 655–662. Role of 4-hydroxy-2,3-nonenal in the pathogenesis of fibrosis. Biofactors 24,
[34] Funes, F. R., Silva, R. C., Arroyo, P. C. Jr., Duca, W. J., Silva, A. A., et al. 229–236.
(2012) Mortality and complications in patients with portal hypertension who [56] Dianzani, M. U., Barrera, G., and Parola, M. (1999) 4-Hydroxy-2,3-nonenal as
underwent transjugular intrahepatic portosystemic shunt (TIPS)—12 years a signal for cell function and differentiation. Acta Biochim. Pol. 46, 61–75.
experience. Arq. Gastroenterol. 49, 143–149. [57] Fernandez, M., Semela, D., Bruix, J., Colle, I., Pinzani, M., et al. (2009)
[35] Kershenobich Stalnikowitz, D. and Weissbrod, A. B (2003) Liver fibrosis and Angiogenesis in liver disease. J. Hepatol. 50, 604–620.
inflammation. A review. Ann. Hepatol. 2, 159–163. [58] Chaisson, M. L., Brooling, J. T., Ladiges, W., Tsai, S., and Fausto, N. (2002)
[36] Vera-Ramirez, L., Sanchez-Rovira, P., Ramirez-Tortosa, M. C., Ramirez-Tor- Hepatocyte specific inhibition of NF-kappaB leads to apoptosis after TNF
tosa, C. L., Granados-Principal, S., et al. (2011) Free radicals in breast carci- treatment, but not after partial hepatectomy. J. Clin. Invest. 110, 193–202.
nogenesis, breast cancer progression and cancer stem cells. Biological [59] Yuan, L. and Kaplowitz, N. (2009) Glutathione in liver diseases and hepato-
bases to develop oxidative-based therapies. Crit. Rev. Oncol. Hematol. 80, toxicity. Mol. Aspects Med. 30, 29–41.
347–368. [60] Diogo, C. V., Grattagliano, I., Oliveira, P. J., Bonfrate, L., and Portincasa, P.
[37] Novo, E. and Parola, M. (2008) Redox mechanisms in hepatic chronic (2011) Re-wiring the circuit, mitochondria as a pharmacological target in
wound healing and fibrogenesis. Fibrogenesis Tissue Repair 13, 5. liver disease. Curr. Med. Chem. 18, 5448–5465.
[38] Holt, A. P., Salmon, M., Buckley, C. D., and Adams, D. H. (2008) Immune [61] Micheau, O. and Tschopp, J. (2003) Induction of TNF receptor I-mediated
interactions in hepatic fibrosis. Clin. Liver Dis. 12, 861–882. apoptosis via two sequential signaling complexes. Cell 114, 181–190.
[39] Consolo, M., Amoroso, A., Spandidos, D. A., and Mazzarino, M. C. (2009) [62] Vandenabeele, P., Declercq, W., Van Herreweghe, F., and Vanden Berghe,
Matrix metalloproteinases and their inhibitors as markers of inflammation T. (2010) The role of the kinases RIP1 and RIP3 in TNF-induced necrosis.
and fibrosis in chronic liver disease. Int. J. Mol. Med. 24, 143–152. Sci. Signal. 3, 4.
[40] Paternostro, C., David, E., Novo, E., and Parola, M. (2010) Hypoxia, angio- [63] Parsons, C. J., Takashima, M., and Rippe, R. A. (2007) Molecular mecha-
genesis and liver fibrogenesis in the progression of chronic liver diseases. nisms of hepatic fibrogenesis. J. Gastroenterol. Hepatol. 22, 79–84.
World J. Gastroenterol. 16, 281–288. [64] Garcı́a-Trevijano, E. R., Iraburu, M. J., Fontana, L., Domı́nguez-Rosales, J.
[41] Parsons, M. J. and Green, D. R. (2011) Death receptors and mitochondria, A., Auster, A., et al. (1999) Transforming growth factor beta1 induces the
life depends on the liver. Hepatology 54, 13–15. expression of alpha1(I) procollagen mRNA by a hydrogen peroxide-C/

98 Curcumin and Liver Disease


EBPbeta-dependent mechanism in rat hepatic stellate cells. Hepatology 29, [83] Lukita-Atmadja, W., Ito, Y., Baker, G. L., and McCuskey, R. S. (2002) Effect
960–970. of curcuminoids as anti-inflammatory agents on the hepatic microvascular
[65] Gressner, O. A., Lahme, B., Demirci, I., Gressner, A. M., and Weiskirchen, R. response to endotoxin. Shock 17, 399–403.
(2007) Differential effects of TGF-beta on connective tissue growth factor [84] Kaur, G., Tirkey, N., Bharrhan, S., Chanana, V., Rishi, P., et al. (2006) Inhibi-
(CTGF/CCN2) expression in hepatic stellate cells and hepatocytes. J. Hepa- tion of oxidative stress and cytokine activity by curcumin in amelioration of
tol. 47, 699–710. endotoxin-induced experimental hepatoxicity in rodents. Clin. Exp. Immu-
[66] Paradis, V., Dargere, D., Bonvoust, F., Vidaud, M., Segarini, P., et al. (2002) nol. 145, 313–321.
Effects and regulation of connective tissue growth factor on hepatic stellate [85] Yun, S. S., Kim, S. P., Kang, M. Y., and Nam, S. H. (2010) Inhibitory effect
cells. Lab. Invest. 82, 767–774. of curcumin on liver injury in a murine model of endotoxemic shock. Bio-
[67] Iizuka, M., Murata, T., Hori, M., and Ozaki, H. (2011) Increased contractility technol. Lett. 32, 209–214.
of hepatic stellate cells in cirrhosis is mediated by enhanced Ca2þ-depend- ňova
[86] Cerný, D., Lekić, N., Va , K., Muchova
, L., Hořı́nek, A., et al. (2011) Hepa-
ent and Ca2þ-sensitization pathways. Am. J. Physiol. Gastrointest. Liver toprotective effect of curcumin in lipopolysaccharide/-galactosamine model
Physiol. 300, 1010–1021. of liver injury in rats: relationship to HO-1/CO antioxidant system. Fitotera-
[68] Rhee, S. G. (2006) Cell signaling. H2O2, a necessary evil for cell signaling. pia 82, 786–791.
Science 12, 1882–1883. [87] Kim, K. M., Pae, H. O., Zhung, M., Ha, H. Y., Ha, Y. A., et al. (2008) Involve-
[69] Boivin, B., Zhang, S., Arbiser, J. L., Zhang, Z. Y., and Tonks, N. K. (2008) A ment of anti-inflammatory heme oxygenase-1 in the inhibitory effect of cur-
modified cysteinyl-labeling assay reveals reversible oxidation of protein ty- cumin on the expression of pro-inflammatory inducible nitric oxide
rosine phosphatases in angiomyolipoma cells. Proc. Natl. Acad. Sci. USA synthase in RAW264.7 macrophages. Biomed. Pharmacother. 62, 630–636.
105, 9959–9964. [88] Liang, G., Li, X., Chen, L., Yang, S., Wu, X., et al. (2008) Synthesis and anti-
[70] Leclercq, I. A., Farrell, G. C., Field, J., Bell, D. R., Gonzalez, F. J., et al. (2001) inflammatory activities of mono-carbonyl analogues of curcumin. Bioorg.
CYP2E1 and CYP4A as microsomal catalysts of lipid peroxides in murine Med. Chem. Lett. 18, 1525–1529.
nonalcoholic steatohepatitis. J. Clin. Invest. 105, 1067–1075. [89] Weisberg, S. P., Leibel, R., and Tortoriello, D. V. (2008) Dietary curcumin
[71] Kathirvel, E., Morgan, K., French, S. W., and Morgan, T. R. (2009) Overex- significantly improves obesity-associated inflammation and diabetes in
pression of liver-specific cytochrome P4502E1 impairs hepatic insulin sig- mouse models of diabesity. Endocrinology 149, 3549–2558.
naling in a transgenic mouse model of nonalcoholic fatty liver disease. Eur. [90] Samuhasaneeto, S., Thong-Ngam, D., Kulaputana, O., Suyasunanont, D.,
J. Gastroenterol. Hepatol. 21, 973–983. and Klaikeaw, N. (2009) Curcumin decreased oxidative stress, inhibited NF-
[72] Chanse aume, E., Tardy, A. L., Salles, J., Giraudet, C., Rousset, P., et al. kappaB activation, and improved liver pathology in ethanol-induced liver
(2007) Chronological approach of diet-induced alterations in muscle mito- injury in rats. J. Biomed. Biotechnol. 2009, 981963.
chondrial functions in rats. Obesity (Silver Spring) 15, 50–59. [91] Shapiro, H., Ashkenazi, M., Weizman, N., Shahmurov, M., Aeed, H., et al.
[73] Laurent, D., Yerby, B., Deacon, R., and Gao, J. (2007) Diet-induced modula- (2006) Curcumin ameliorates acute thioacetamide-induced hepatotoxicity. J.
tion of mitochondrial activity in rat muscle. Am. J. Physiol. Endocrinol. Gastroenterol. Hepatol. 21, 358–366.
Metab. 293, 1169–1177. [92] Bisht, S., Khan, M. A., Bekhit, M., Bai, H., Cornish, T., et al. (2011) A poly-
[74] Krauss, S., Zhang, C. Y., Scorrano, L., Dalgaard, L. T., St-Pierre, J., et al. meric nanoparticle formulation of curcumin (NanoCurcTM) ameliorates
(2003) Superoxide mediated activation of uncoupling protein 2 causes pan- CCl4-induced hepatic injury and fibrosis through reduction of pro-inflam-
creatic beta cell dysfunction. J. Clin. Invest. 112, 1831–1842. matory cytokines and stellate cell activation. Lab. Invest. 91, 1383–1395.
[75] Couplan, E., del Mar Gonzalez-Barroso, M., Alves-Guerra, M. C., Ricquier, [93] Leclercq, I. A., Farrell, G. C., Sempoux, C., de la Pen ~ a, A., and Horsmans, Y.
D., Goubern, M., et al. (2002) No evidence for a basal, retinoic, or superox- (2004) Curcumin inhibits NF-kappaB activation and reduces the severity of
ide induced uncoupling activity of the uncoupling protein 2 present in experimental steatohepatitis in mice. J. Hepatol. 41, 926–934.
spleen or lung mitochondria. J. Biol. Chem. 277, 26268–26275. [94] Charoensuk, L., Pinlaor, P., Prakobwong, S., Hiraku, Y., Laothong, U., et al.
[76] Silva, J. P., Shabalina, I. G., Dufour, E., Petrovic, N., Backlund, E. C., et al. (2011) Curcumin induces a nuclear factor-erythroid 2-related factor 2-driven
(2005) SOD2 overexpression, enhanced mitochondrial tolerance but ab- response against oxidative and nitrative stress after praziquantel treatment
sence of effect on UCP activity. EMBO J. 24, 4061–4070. in liver fluke-infected hamsters. Int. J. Parasitol. 41, 615–626.
[77] Schreurs, M., Kuipers, F., and van der Leij, F. R. (2010) Regulatory enzymes [95] Mouzaoui, S., Rahim, I., and Djerdjouri, B. (2012) Aminoguanidine and cur-
of mitochondrial beta-oxidation as targets for treatment of the metabolic cumin attenuated tumor necrosis factor (TNF)-a-induced oxidative stress,
syndrome. Obes. Rev. 11, 380–388. colitis and hepatotoxicity in mice. Int. Immunopharmacol. 12, 302–311.
[78] Memon, R. A., Tecott, L. H., Nonogaki, K., Beigneux, A., Moser, A. H., et al. [96] Nanji, A. A., Jokelainen, K., Tipoe, G. L., Rahemtulla, A., Thomas, P., et al.
(2000) Up-regulation of peroxisome proliferator-activated receptors (PPAR- (2003) Curcumin prevents alcohol-induced liver disease in rats by inhibiting
alpha) and PPAR-gamma messenger ribonucleic acid expression in the liver the expression of NF-kappa B-dependent genes. Am. J. Physiol. Gastroint-
in murine obesity, troglitazone induces expression of PPAR-gamma respon- est. Liver Physiol. 284, 321–327.
sive adipose tissue-specific genes in the liver of obese diabetic mice. Endo- [97] Bassiouny, A. R., Zaky, A., and Kandeelm K. M. (2011) Alteration of AP-en-
crinology 141, 4021–4031. donuclease1 expression in curcumin-treated fibrotic rats. Ann. Hepatol. 10,
[79] Gupta, S. C., Patchva, S., Koh, W., and Aggarwal, B. B. (2012) Discovery of 516–530.
curcumin, a component of golden spice, and its miraculous biological activ- [98] Reyes-Gordillo, K., Segovia, J., Shibayama, M., Tsutsumi, V., Vergara, P.,
ities. Clin. Exp. Pharmacol. Physiol. 39, 283–299. et al. (2008) Curcumin prevents and reverses cirrhosis induced by bile duct
[80] Ghosh, N., Ghosh, R., Mandal, V., and Mandal, S. C. (2011) Recent advances obstruction or CCl4 in rats: role of TGF-beta modulation and oxidative
in herbal medicine for treatment of liver diseases. Pharm. Biol. 49, 970–988. stress. Fundam. Clin. Pharm. 22, 417–427.
[81] Matsuda, H., Ninomiya, K., Morikawa, T., and Yoshikawa, M. (1998) [99] Lin, Y. L., Lin, C. Y., Chi, C. W., and Huang, Y. T. (2009) Study on antifibrotic
Inhibitory effect and action mechanism of sesquiterpenes from Zedoariae effects of curcumin in rat hepatic stellate cells. Phytother. Res. 23, 927–932.
Rhizoma on D-galactosamine/lipopolysaccharide-induced liver injury. Bio- [100] Shu, J. C., He, Y. J., Lv, X., Ye, G. R., and Wang, L. X. (2009) Curcumin
org. Med. Chem. Lett. 17, 339–344. prevents liver fibrosis by inducing apoptosis and suppressing activation of
[82] Morikawa, T., Matsuda, H., Ninomiya, K., and Yoshikawa, M. (2002) Medici- hepatic stellate cells. J. Nat. Med. 63, 415–420.
nal foodstuffs. XXIX. Potent protective effects of sesquiterpenes and curcu- [101] Nakayama, N., Nakamura, T., Okada, H., Iwaki, S., Sobel, B. E., et al. (2011)
min from Zedoariae Rhizoma on liver injury induced by D-galactosamine/ Modulators of induction of plasminogen activator inhibitor type-1 in
lipopolysaccharide or tumor necrosis factor-alpha. Biol. Pharm. Bull. 25, HepG2 cells by transforming growth factor-b. Coronary Artery Dis. 22,
627–631. 468–478.

Vera-Ramirez et al. 99
BioFactors

[102] Tu, C. T., Han, B., Yao, Q. Y., Zhang, Y. A., Liu, H. C., et al. (2012) Curcu- demia and hepatic steatosis in fructose-fed rats. Hepatology 51,
min attenuates Concanavalin A-induced liver injury in mice by inhibition 1555–1566.
of Toll-like receptor (TLR) 2, TLR4 and TLR9 expression. Int. Immunophar- [120] Kang, Q. and Chen, A. (2009) Curcumin eliminates oxidized LDL roles in
macol. 12, 151–157. activating hepatic stellate cells by suppressing gene expression of lectin-
[103] Tu, C. T., Yao, Q. Y., Xu, B. L., Wang, J. Y., Zhou, C. H., et al. (2012) Protec- like oxidized LDL receptor-1. Lab. Invest. 89, 1275–1290.
tive effects of curcumin against hepatic fibrosis induced by carbon tetra- [121] Guangwei, X., Rongzhu, L., Wenrong, X., Suhua, W., Xiaowu, Z., et al.
chloride: modulation of high-mobility group box 1, Toll-like receptor 4 and (2010) Curcumin pretreatment protects against acute acrylonitrile-induced
2 expression. Food Chem. Toxicol. 50, 3343–3351. oxidative damage in rats. Toxicology 267, 140–146.
[104] Park, E. J., Jeon, C. H., Ko, G., Kim, J., and Sohn, D. H. (2000) Protective [122] Farombi, E. O., Shrotriya, S., Na, H. K., Kim, S. H., and Surh, Y. J. (2008)
effect of curcumin in rat liver injury induced by carbon tetrachloride. J. Curcumin attenuates dimethylnitrosamine-induced liver injury in rats
Pharm. Pharmacol. 52, 437–440. through Nrf2-mediated induction of heme oxygenase-1. Food Chem. Toxi-
[105] Wu, S. J., Lin, Y. H., Chu, C. C., Tsai, Y. H., and Chao, J. C. (2008) Curcu- col. 46, 1279–1287.
min or saikosaponin a improves hepatic antioxidant capacity and protects [123] Lin, J. and Chen, A. (2008) Activation of peroxisome proliferator-activated
against CCl4-induced liver injury in rats. J. Med. Food 11, 224–229. receptor-gamma by curcumin blocks the signaling pathways for PDGF and
[106] Pari, L. and Murugan, P. (2004) Protective role of tetrahydrocurcumin EGF in hepatic stellate cells. Lab. Invest. 88, 529–540.
against erythromycin estolate-induced hepatotoxicity. Pharmacol. Res. 49, [124] Kuo, J. J., Chang, H. H., Tsai, T. H., and Lee, T. Y. (2012) Positive effect of
481–486. curcumin on inflammation and mitochondrial dysfunction in obese mice
[107] Pari, L. and Amali, R. (2005) Protective role of tetrahydrocurcumin (THC) with liver steatosis. Int. J. Mol. Med. 30, 673–679.
an active principle of turmeric on chloroquine induced hepatotoxicity in [125] He, S. M., Chan, E., and Zhou, S. F. (2011) ADME properties of herbal
rats. J. Pharm. Pharm. Sci. 8, 115–123. medicines in humans: evidence, challenges and strategies. Curr. Pharm.
[108] Naik, S. R., Thakare, V. N., and Patil, S. R. (2011) Protective effect of curcumin Des. 17, 357–407.
on experimentally induced inflammation, hepatotoxicity and cardiotoxicity in [126] Sharma, R. A., Steward, W. P., and Gescher, A. J. (2007) Pharmacokinetics
rats, evidence of its antioxidant property. Exp. Toxicol. Pathol. 63, 419–431. and pharmacodynamics of curcumin. Adv. Exp. Med. Biol. 595, 453–470.
[109] Rong, S., Zhao, Y., Bao, W., Xiao, X., Wang, D., et al. (2012) Curcumin pre- [127] Cheng, A. L., Hsu, C. H., Lin, J. K., Hsu, M. M., Ho, Y. F., et al. (2001) Phase
vents chronic alcohol-induced liver disease involving decreasing ROS gen- I clinical trial of curcumin, a chemopreventive agent, in patients with high-
eration and enhancing antioxidative capacity. Phytomedicine 19, 545–550. risk or pre-malignant lesions. Anticancer Res. 21, 2895–2900.
[110] Eybl, V., Kotyzova, D., and Koutensky, J. (2006) Comparative study of natu- [128] Shoba, G., Joy, D., Joseph, T., Majeed, M., Rajendran, R., et al. (1998)
ral antioxidants - curcumin, resveratrol and melatonin - in cadmium- Influence of piperine on the pharmacokinetics of curcumin in animals and
induced oxidative damage in mice. Toxicology 225, 150–156. human volunteers. Planta Med. 64, 353–356.
[111] Girish, C., Koner, B. C., Jayanthi, S., Ramachandra, R. K., Rajesh, B., et al. [129] Kanai, M., Imaizumi, A., Otsuka, Y., Sasaki, H., Hashiguchi, M., et al. (2012)
(2009) Hepatoprotective activity of picroliv, curcumin and ellagic acid com- Dose-escalation and pharmacokinetic study of nanoparticle curcumin, a
pared to silymarin on paracetamol induced liver toxicity in mice. Fundam. potential anticancer agent with improved bioavailability, in healthy human
Clin. Pharm. 23, 735–745. volunteers. Cancer Chemother. Pharmacol. 69, 65–70.
[112] Yousef, M. I., Omar, S. A., El-Guendi, M. I., and Abdelmegid, L. A. (2010) [130] Maiti, K., Mukherjee, K., Gantait, A., Saha, B. P., and Mukherjee, P. K.
Potential protective effects of quercetin and curcumin on paracetamol- (2007) Curcumin-phospholipid complex: preparation, therapeutic evalua-
induced histological changes, oxidative stress, impaired liver and kidney tion and pharmacokinetic study in rats. Int. J. Pharm. 330, 155–163.
functions and haematotoxicity in rat. Food Chem. Toxicol. 48, 3246–3261. [131] Lao, C. D., Ruffin, M. T. IV, Normolle, D., Heath, D. D., Murray, S. I., et al.
[113] Rao, D. S., Sekhara, N. C., Satyanarayana, M. N., and Srinivasan, M. (2006) Dose escalation of a curcuminoid formulation. BMC Complement
(1970) Effect of curcumin on serum and liver cholesterol levels in the rat. Altern. Med. 6, 10.
J. Nutr. 100, 1307–1315. [132] Balaji, S. and Chempakam, B. (2010) Toxicity prediction of compounds
[114] Asai, A. and Miyazawa, T. (2001) Dietary curcuminoids prevent high-fat from turmeric (Curcuma longa L). Food Chem. Toxicol. 48, 2951–2959.
diet-induced lipid accumulation in rat liver and epididymal adipose tissue. [133] Quiles, J. L., Huertas, J. R., Man ~ as, M., Ochoa, J. J., Battino, M., et al.
J. Nutr. 131, 2932–2935. (1999) Oxidative stress induced by exercise and dietary fat modulates the
[115] Rukkumani, R., Sri Balasubashini, M., Vishwanathan, P., and Menon, V. P. coenzyme Q and vitamin A balance between plasma and mitochondria.
(2002) Comparative effects of curcumin and photo-irradiated curcumin on Int. J. Vitam. Nutr. Res. 69, 243–249.
alcohol- and polyunsaturated fatty acid-induced hyperlipidemia. Pharma- [134] Battino, M., Quiles, J. L., Huertas, J. R., Ramirez-Tortosa, M. C., Cassinello,
col. Res. 46, 257–264. M., et al. (2002) Feeding fried oil changes antioxidant and fatty acid pat-
[116] Rukkumani, R., Aruna, K., Varma, P. S., Viswanathan, P., Rajasekaran, K. tern of rat and affects rat liver mitochondrial respiratory chain compo-
N., et al. (2005) Protective role of a novel curcuminoid on alcohol and nents. J. Bioenerg. Biomembr. 34, 127–134.
PUFA-Induced hyperlipidemia. Toxicol. Mech. Methods 15, 227–234. [135] Quiles, J. L., Huertas, J. R., Battino, M., Ramı́rez-Tortosa, M. C., Cassinello,
[117] Jang, E. M., Choi, M. S., Jung, U. J., Kim, M. J., Kim, H. J., et al (2008) M., et al. (2002) The intake of fried virgin olive or sunflower oils differen-
Beneficial effects of curcumin on hyperlipidemia and insulin resistance in tially induces oxidative stress in rat liver microsomes. Br. J. Nutr. 88,
high-fat-fed hamsters. Metabolism 57, 1576–1583. 57–65.
[118] Shao, W., Yu, Z., Chiang, Y., Yang, Y., Chai, T., et al. (2012) Curcumin pre- [136] Ochoa, J. J., Quiles, J. L., Iba n
~ ez, S., Martı́nez, E., Lo
 pez-Frı́as, M., et al.
vents high fat diet induced insulin resistance and obesity via attenuating (2003) Aging-related oxidative stress depends on dietary lipid source in rat
lipogenesis in liver and inflammatory pathway in adipocytes. PLoS One 7, postmitotic tissues. J. Bioenerg. Biomembr. 35, 267–275.
28784. [137] Mancuso, M., Orsucci, D., Filosto, M., Simoncini, C., and Siciliano, G.
[119] Li, J. M., Li, Y. C., Kong, L. D., and Hu, Q. H. (2010) Curcumin inhibits (2012) Drugs and mitochondrial diseases: 40 queries and answers. Expert
hepatic protein-tyrosine phosphatase 1B and prevents hypertriglyceri- Opin. Pharmacother. 13, 527–543.

100 Curcumin and Liver Disease

You might also like