ArticlePDF AvailableLiterature Review

Curcumin and cellular stress response in free radical-related diseases

Authors:

Abstract and Figures

Free radicals play a main pathogenic role in several human diseases such as neurodegenerative disorders, diabetes, and cancer. Although there has been progress in treatment of these diseases, the development of important side effects may complicate the therapeutic course. Curcumin, a well known spice commonly used in India to make foods colored and flavored, is also used in traditional medicine to treat mild or moderate human diseases. In the recent years, a growing body of literature has unraveled the antioxidant, anticarcinogenic, and antinfectious activity of curcumin based on the ability of this compound to regulate a number of cellular signal transduction pathways. These promising data obtained in vitro are now being translated to the clinic and more than ten clinical trials are currently ongoing worldwide. This review outlines the biological activities of curcumin and discusses its potential use in the prevention and treatment of human diseases.
Content may be subject to copyright.
1062
Review
Curcumin and the cellular stress response in free
radical-related diseases
Vittorio Calabrese1, Timothy E. Bates2, Cesare Mancuso3, Carolin Cornelius1,
Bernardo Ventimiglia4, Maria Teresa Cambria1, Laura Di Renzo5, Antonino De Lorenzo5
and Albena T. Dinkova-Kostova6, 7
1Department of Chemistry, Clinical Biochemistry and Clinical Molecular Biology Chair, University of
Catania, Catania, Italy
2School of Biomedical Sciences, University of Nottingham, Nottingham, UK
3Institute of Pharmacology, Catholic University School of Medicine, Roma, Italy
4Department of Science of Senescence, Urology and Neuro-Urology, University of Catania, Italy
5Human Nutrition Unit, Department of Neuroscience, University of Rome Tor Vergata, Rome, Italy
6Division of Clinical Pharmacology, Departments of Medicine and Pharmacology and Molecular Sciences,
Johns Hopkins University School of Medicine, Baltimore, MD, USA
7Biomedical Research Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
Free radicals play a main pathogenic role in several human diseases such as neurodegenerative disor-
ders, diabetes, and cancer. Although there has been progress in treatment of these diseases, the devel-
opment of important side effects may complicate the therapeutic course. Curcumin, a well known
spice commonly used in India to make foods colored and flavored, is also used in traditional medicine
to treat mild or moderate human diseases. In the recent years, a growing body of literature has unrav-
eled the antioxidant, anticarcinogenic, and antinfectious activity of curcumin based on the ability of
this compound to regulate a number of cellular signal transduction pathways. These promising data
obtained in vitro are now being translated to the clinic and more than ten clinical trials are currently
ongoing worldwide. This review outlines the biological activities of curcumin and discusses its poten-
tial use in the prevention and treatment of human diseases.
Keywords: Curcumin / Free radicals / Heat shock proteins / Heme oxygenase / Neurodegenerative disorders /
Received: August 10, 2007; revised: April 29, 2008; accepted: May 1, 2008
1 Introduction
Curcumin (1,7-bis(4-hydroxy 3-methoxy phenyl)-1,6-hep-
tadiene-3,5-dione) (Fig. 1) is a phenolic compound
extracted from the rhizome of Curcuma longa L. (family
Zingiberaceae) and it is commonly used in the Asian conti-
nent, especially in India, as a spice to make food colored
and flavored. Furthermore, traditional Indian medicine has
considered curcumin a drug effective on several disorders
including anorexia, coryza, cough, hepatic diseases, and
sinusitis [1, 2]. Recently, several studies have substantiated
and provided scientific evidence regarding the potential
prophylactic or therapeutic use of curcumin, unraveling the
anti-inflammatory, anticarcinogenic, and anti-infectious
activities of this compound [3–6]. In the field of neuropro-
tection, especially exciting are the findings of Cole and his
colleagues who demonstrated that curcumin inhibits forma-
tion of amyloid boligomers and fibrils at submicromolar
concentrations, crosses the blood–brain barrier, and
reduces amyloid levels and plaque burden in a mouse model
of Alzheimer's disease (AD) [7].
2 Pharmacokinetics of curcumin
Curcumin is quite stable at acidic pH and upon ingestion
almost 40–80% of this compound remains in the gastroin-
Correspondence: Professor Vittorio Calabrese, Department of Chem-
istry, Biochemistry and Molecular Biology Section, University of Cat-
ania, Via Andrea Doria 95100 Catania, Italy
E-mail: calabres@mbox.unict.it
Fax: +39-095-580138
Abbreviations: AD, Alzheimer's disease; APP, amyloid precursor
protein; ARE, antioxidant response element; BM, bone marrow; BR,
bilirubin; BV, biliverdin; GST, glutathione S-transferase; HIF-1, hypo-
xia-inducible factor-1; HO-1, heme oxygenase-1; Hsp70, heat shock
protein70; IkB, inhibitory kappa B; Keap1, Kelch-like ECH-associ-
ated protein 1; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine;
NFkB, nuclear factor kB; NO, nitric oxide; NOS, nitric oxide syn-
thase; Nrf2, nuclear factor-erythroid 2-related factor 2; PD, Parkin-
son's disease; RNS, reactive nitrogen species; ROS, reactive oxygen
species; THC, tetrahydrocurcumin; Trx, thioredoxin; TrxR, thiore-
doxin reductase; UPR, unfolded protein response; UPS, ubiquitin
proteasome system
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
DOI 10.1002/mnfr.200700316 Mol. Nutr. Food Res. 2008, 52, 1062 –1073
Mol. Nutr. Food Res. 2008, 52, 1062 1073
testinal tract [1]. However, curcumin undergoes a marked
first-pass metabolism which limits its systemic bioavail-
ability (l60%) as demonstrated in humans and rodents [8
10]. Interestingly, in order to increase its bioavailability, the
co-administration of curcumin with piperine or its com-
plexation with phospholipids to form a curcumin–phos-
pholipid complex have been proposed [8, 11, 12]. Preclini-
cal studies have shown that administration of 1 g/kg of cur-
cumin to the rat allows the polyphenol to reach plasma con-
centrations around 0.5 lg/mL; on the other hand, patients
affected by malignant or premalignant conditions of the
bladder, skin, cervix, stomach, or oral mucosa, treated with
doses of curcumin in the range of 0.5–8 g/day for 3 months
had a plasma concentration of this compound of
1.75 l0.8 lM [8, 13]. In the rat, the volume of distribution
of curcumin is around 190 L suggesting that this polyphe-
nol may accumulate in many organs including colorectal
tissue, liver, and brain [8, 11, 14]. Studies in rodents and
humans demonstrated that, after oral dosing, curcumin is
conjugated to curcumin glucuronide and curcumin sulfate
as well as reduced into dihydrocurcumin (DHC), tetrahy-
drocurcumin (THC), hexahydrocurcumin, octahydrocurcu-
min, and hexahydrocurcuminol [1, 15, 16]; curcumin,
DHC, and THC can be further converted in monoglucuro-
nide conjugates [15, 17]. These metabolic changes seem to
occur not only in the liver, the main organ deputed to bio-
transformation, but also in the intestinal tract [1, 16]. Inter-
estingly, the metabolism of curcumin generates products
such as THC which retains anti-inflammatory activity com-
parable to that of the parental compound [1, 16]. In rodents
and humans curcumin inhibits cytochrome P450 enzymes,
glutathione S-transferase (GST) and UDP-glucuronosyl-
transferases, therefore the ingestion of this spice may alter
the metabolism of drugs thus increasing their plasma con-
centrations and initiating potential toxic effects [18–21]. In
the rat, curcumin is mainly excreted into the bile and elimi-
nated in the feces, only a small amount is excreted in the
urine [9, 10] with a half-life of elimination of l1.5 h [11].
The urinary elimination of curcumin and its metabolites
seems to increase if curcumin is administered at large doses
(e.g., 3.6 g/day for up to 4 months) [8, 22]. With regard to
the toxicity profile of curcumin, studies in rodents and pri-
mates have shown that doses of up to 3.5 g/kg body weight
administered for up to 3 months were well tolerated by the
animals [8]. In humans, curcumin at doses ranging from 2.1
to 8 g/day for up to 3 months did not originate any toxic
effects [13, 23]. However, patients affected by advanced
colorectal cancer treated with curcumin (3.6 g/day) devel-
oped diarrhea whereas a dose of 0.9 g/day was associated
with nausea, which resolved spontaneously. In the same
patients, blood test abnormalities related to curcumin
administration were a rise in serum alkaline phosphatase
and lactate dehydrogenase, but the possibility that they
resulted from the progression of cancer rather than curcu-
min toxicity can not be excluded [8, 22].
3 Pharmacodynamics of curcumin
Early studies have shown that curcumin and related prod-
ucts such as THC, have a strong antioxidant activity. In fact,
these compounds have been shown to reduce free radical-
or copper-induced lipid peroxidation in several experimen-
tal systems [24–26]. Furthermore, structure activity stud-
ies clearly demonstrated the importance of the b-diketone
moiety and especially the phenolic hydroxyl group, for the
antioxidant activity of curcumin and its analogues [24, 27].
Very recently, many papers have appeared in the literature
demonstrating that curcumin and its metabolites affect
numerous intracellular systems such as transcription factor
nuclear factor kB (NFkB), inducible nitric oxide synthase
(iNOS), hypoxia-inducible factor-1 (HIF-1), nuclear factor-
erythroid 2-related factor 2 (Nrf2), and members of the vita-
gene family (e.g., heat shock protein70 (Hsp70), heme oxy-
genase-1 (HO-1), thioredoxin (Trx)). This complex array of
interactions is in agreement with the well-known ability of
curcumin to serve not only as an antioxidant but also as
anti-inflammatory and anticarcinogenic molecule.
Reyes-Gordillo et al. [28], in an elegant paper, have shown
that curcumin reduced the CCl4-induced liver toxicity in the
rat; in particular, curcumin reduced the CCl4-related
increase in proinflammatory cytokines and blocked the
nuclear translocation of NFkB [28]. Similarly, curcumin pre-
vented the dinitrochlorobenzene-induced colitis in the rat by
downregulating both NFkB and iNOS [29]. In lung epithelial
cells, curcumin exerted anticarcinogenic activity and pre-
vented the cigarette smoke-induced NFkB activation
through the inhibition of inhibitory kappa B (IkB)akinase
activation, IkBaphosphorylation, and degradation [30]. The
inhibition of the NFkB activation was paralleled bythe sup-
pression of many NFkB-related genes, including cyclin D1,
cyclooxygenase-2, and matrix metalloproteinase-9 (MMP-
9) [30]. Comparable results have been found in a macro-
phage cell line (RAW 264.7) challenged with bacterial endo-
toxin. In these cells, curcumin and its reduced metabolites
blocked the activation of NFkB, and the downstream activa-
tion of iNOS, via inhibition of the IkB kinases 1 and 2, thus
providing further evidence about the importance of the
effects on NFkB in the anti-inflammatory and anticarcino-
genic activity of this phenolic compound [31]. Through
interaction with NFkB, curcumin exerts protective function
also in the regulation of T-cell-mediated immunity. In fact,
overexpression of NFkB in T-cells confers protection against
tumor-induced apoptosis, whereas when NFkB is inhibited,
the cell becomes much more vulnerable and undergoes apop-
tosis [32]. By so doing, NFkB plays an important role in the
regulation of T-cell apoptosis and the related thymic atrophy
which occurs during carcinogenesis. In this experimental
model, curcumin prevented the tumor-induced apoptosis
and the following thymic atrophy by restoring the activity of
NFkB [32].
1063
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
V. Calabrese et al. Mol. Nutr. Food Res. 2008, 52, 1062 –1073
Another transcription factor involved in the anticarcino-
genic effect of curcumin is HIF-1. HIF-1 is composed of
two proteins, HIF-1aand the aryl hydrocarbon receptor
nuclear translocator (ARNT) and plays a major role in the
development of hypoxic tumors [33]. Curcumin has been
demonstrated to inactivate HIF-1 in several cell lines and
this effect has been related to its ability to promote ARNT
degradation [33]. As a consequence of HIF-1 inactivation,
several proteins downstream to HIF-1 were downregulated,
such as erythropoietin and the vascular endothelial growth
factor (VEGF) [33].
Particularly interesting is the interaction of curcumin
with the vitagene system. The term vitagenes refers to an
integrated network of protective mechanisms involved in
preserving cellular homeostasis during stress conditions,
which are under control of redox-sensitive genes and
related signaling pathways that result in increased expres-
sion of specific genes, such as those responding to antioxi-
dant compounds [34 36]. The vitagene family is composed
of the Hsps HO-1, Hsp70 and by the Trx/thioredoxin reduc-
tase (TrxR) system [3436]. HO-1, also referred to as
Hsp32, degrades heme, which is toxic if produced in excess,
into ferrous iron, carbon monoxide, and biliverdin (BV);
BV is the precursor of bilirubin (BR), a linear tetrapyrrole
which has been shown to effectively counteract oxidative
and nitrosative stress due to its ability to interact with reac-
tive oxygen species (ROS), nitric oxide (NO), and reactive
nitrogen species (RNS) [34, 35, 3740]. Hsp70 is a func-
tional chaperone and acts by inhibiting key effectors of the
apoptotic machinery [34, 37]. Finally, Trx is responsible for
the reduction of protein disulfide bonds whereas TrxR
serves to maintain Trx in a reduced form [34]. Very
recently, curcumin was shown to have cytoprotective effects
by interacting with all members of the vitagene family. In
particular, curcumin increased the expression of HO-1 in
human cardiac myoblasts, hepatocytes, monocytes, and
endothelial cells [41 44], rat neurons and astrocytes [45]
as well as porcine endothelial cells [46]. In several rodents
and human cells, the curcumin-induced HO-1 overexpres-
sion was correlated with production of mitochondrial ROS,
activation of transcription factors Nrf2 and NFkB, induc-
tion of mytogen activated protein kinase (MAPK) p38, and
inhibition of phosphatase activity [44, 47, 48]. Moreover,
curcumin upregulated Hsp70 in human colorectal carci-
noma cells, proximal tubule cells [4952], and rat glioma
cells [53]. Quite different is the effect of curcumin on TrxR,
as it has been shown that curcumin irreversibly inhibits
TrxR activity. As a consequence, there was increased
NADPH oxidase activity, which in turn, produced an abun-
dance of ROS [54]. This latter paradoxical effect may
explain, at least in part, the cancer chemopreventive activity
of curcumin [54].
Having two Michael acceptor functionalities on its mole-
cule (Fig. 1), curcumin and its structural analogues induce
the gene expression of a battery of cytoprotective proteins
in a process known as “the phase 2 response” [55] or the
“electrophile counterattack response” [56]. The designation
“phase 2” historically comes from the fact that many of
these proteins are involved in the second step of the metab-
olism of xenobiotics, e.g., the diverse families of gluta-
thione transferases and UDP-glucuronosyltransferases
[57]. However, the understanding of the common molecular
regulation of the basal and inducible levels of their gene
expression has placed many other proteins, e.g., HO-1 and
c-glutamylcysteine synthetase, in this category of cytopro-
tective proteins [58, 59]. There are three essential cellular
components in the general scheme of induction of the phase
2 response (Fig. 2): (i) the antioxidant response elements
(AREs), (ii) transcription factor Nrf2, and (iii) the sensor
for inducers Kelch-like ECH-associated protein 1 (Keap1).
The AREs represent single or multiple copies of upstream
regulatory sequences present on all genes encoding for
phase 2 cytoprotective proteins [60, 61]. Binding to the
AREs is Nrf2, a 66-kDa transcription factor [62], the princi-
pal transcription factor that determines the levels of expres-
sion of these cytoprotective genes [63]. Nrf2 is a cap ,n'-col-
lar (CNC) transcription factor that has a highly conserved
basic region-leucine zipper (bZIP) domain [64]. In order to
bind to the AREs, Nrf2 has to form a heterodimer first with
a small Maf protein [65]. Binding of the dimeric complex
so formed to the ARE and subsequent recruitment of the
general transcriptional machinery ultimately results in the
activation of transcription of ARE-dependent genes and
correlates with protection against a wide array of electro-
philes and oxidants [66 68]. A great body of genetic evi-
dence utilizing nrf2-knockout mice reveled that Nrf2 serves
as a master regulator of the ARE-driven cellular defense
systems against electrophiles and oxidants. Indeed, nrf2-
knockout mice have low and uninducible levels of gluta-
thione and phase 2 proteins compared to otherwise geneti-
1064
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Figure 1. Schematic diagram showing the main molecular tar-
gets of curcumin. Curcumin inhibits pathways that could lead
to neurodegeneration and upregulates cytoprotective (phase
2) enzymes and vitagenes thus counteracting free radical-
induced damage and exerting a neuroprotective role. By mod-
ulating the activation of various transcription factors, curcumin
regulates the expression of inflammatory enzymes, cytokines,
adhesion molecules, and cell survival proteins. Curcumin also
downregulates cyclin D1, COX-2, and MMP-9, and upregu-
lates p21, p27, and p53. Straight arrows, stimulation; dashed
arrows, inhibition.
Mol. Nutr. Food Res. 2008, 52, 1062 1073
cally identical wild-type mice and are much more sensitive
to toxic challenges of many different types [66 68].
The entry of Michael acceptors like curcumin into the
cell is “sensed” by the cysteine-rich sensor protein Keap1
[69], a dimeric cytosolic repressor protein that binds and
targets Nrf2 for ubiquitination and subsequent proteasomal
degradation via association with Cullin 3 to form an E3
ubiquitin ligase complex [70]. Inducers chemically mod-
ify specific highly reactive cysteine residues of Keap1 [71,
72]. Direct sulfhydryl-addition reaction of the structurally
related to curcumin synthetic bis(2-hydroxybenzylidene)a-
cetone with purified recombinant murine Keap1 has been
observed spectroscopically [71]. Such reaction with the
cysteine sulfhydryls of Keap1 leads to loss of its ability to
repress Nrf2 which then undergoes nuclear translocation
and activates the transcription of ARE-dependent genes
[66 73]. Murine Keap1 has 25 cysteine residues (its human
homologue has 27) and amino acid replacements of C273
and C288, either individually or in combination, result in
loss of the repressor function of Keap1 and constitutive
expression of ARE-dependent genes [73].
Notably, the gene expression of HO-1, Trx, and TrxR can
be upregulated in a manner dependent on Nrf2 and ARE.
To our knowledge, there are no reports of a similar type of
regulation of the third member of the vitagene family,
Hsp70. However, curcumin as well as several other com-
pounds (e.g., hydrogen peroxide [74], the cyclopentenone
prostaglandin 15-deoxy-D12,14-prostaglandin J2, and the
vicinal dithiol reagent phenylarsine oxide [75, 76]), all of
which can react with sulfhydryl groups, have been reported
to increase the protein levels of Hsp70. Indeed, the tran-
scriptional activation of both Nrf2 and heat shock factor 1
(HSF1), the major activator of Hsp70 gene expression,
depend on cysteine modification, in one case within the
Nrf2 regulator Keap1 [72, 76], and in the other, within
HSF1 [77].
4 Curcumin and neurodegenerative
disorders
Neurodegenerative disorders, such as AD and Parkinson's
disease (PD), belong to the family of the protein conforma-
tional diseases (PCD) and affect a large portion of our aging
population [78]. In general, PCD are conditions that arise
from the dysfunctional aggregation of proteins in non
native conformations. It is known that the b-conformation
in proteins is particularly susceptible to perturbations in the
quality control system and that ROS play an important role
in the development and/or pathogenetic progression of
aging and neurodegenerative diseases [79 81]. Chaperones
can rescue misfolded proteins by breaking up aggregates
and assisting the refolding process [80, 82]. Proteins that
cannot be rescued by refolding can be delivered to the pro-
teasome by chaperones to be recycled [82, 59]. If the cell is
not able to eliminate misfolded proteins multiple metabolic
derangements resulting in the excessive production of ROS
and RNS occur [83]. The ability of a cell to deal with oxida-
tive and nitrosative stress requires functional chaperones,
antioxidant production, protein degradation, and a cascade
of intracellular events collectively known as unfolded pro-
tein response (UPR), a form of cell stress response [84, 85].
As the cell's quality control system becomes overwhelmed
(see below), conformational changes occur to amyloid pol-
ypeptide intermediates, generating stable oligomers with an
antiparallel crossed b-pleated sheet structure that eventu-
ally accumulate as space-occupying lesions within neurons
[81]. Although it is clear why mutant proteins form amy-
loid, it is hard to rationalize why a wild-type protein adopts
a native conformation in most individuals, but it misfolds in
a minority of others, in what should be a common extracel-
lular environment. This discrepancy suggests that other
events most likely trigger misfolding processes in sporadic
amyloid disease. One possibility is that an abnormal metab-
olite, generated only in some individuals, covalently modi-
fies the protein or peptide and causes it to misfold. Candi-
date metabolites are suggested by the recently recognized
links between AD and atherosclerosis, in which known
1065
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Figure 2. Model for induction of phase 2 cytoprotective genes.
Nrf2 is a transcription factor responsible for the induction of
several genes related to the cellular stress response, including
phase 2 enzymes, such as GST and NAD(P)H:quinone oxidor-
eductase 1 (NQO1); antioxidants, such as glutathione reduc-
tase (GR), glutathione peroxidase (GPx), and catalase; and
vitagenes such as Heme oxygenase-1 (HO-1) and Hsp70. At
basal conditions the cysteine rich sensor metalloprotein
Keap1 binds and targets transcription factor Nrf2 for ubiquiti-
nation and proteasomal degradation via association with the
Cullin 3 (Cul3)-based E3 ubiquitin ligase complex. Inducers,
by promoting mild oxidative stress (hormesis) react and
chemically modify specific highly reactive cysteine residues of
the sensor Keap1. Consequently, Keap1 loses its ability to
repress transcription factor Nrf2. This leads to increased stabi-
lization of Nrf2, its nuclear translocation, binding to the ARE
(in heterodimeric combinations with members of the small Maf
family of transcription factors), and ultimately transcriptional
activation of phase 2 cytoprotective genes.
V. Calabrese et al. Mol. Nutr. Food Res. 2008, 52, 1062 –1073
chronic inflammatory metabolites, may play a critical
pathogenic role. If this holds true, then new targets are dis-
closed for a prevention strategy brought about through
nutritional antioxidants.
AD is characterized by a subtly impaired cognitive func-
tion or a disturbance of behavior. With time there is a grad-
ual memory loss and disorientation which eventually prog-
ress into dementia. Although, most cases are sporadic, 5
10% or more are familial [86]. Families that have an autoso-
mal dominant pattern for AD constitute about 13% of early
cases and less than 0.01% of the total number of patients.
Molecular analysis of families with early onset AD has
made it possible to identify mutations in three different
genes that are responsible for the disease: the gene encoding
for the amyloid precursor protein (APP) peptide, and the
presenilin 1 (PSEN1) and presenilin 2 (PSEN2) genes. Yet,
these genes are involved in less than 5% of the total number
of cases of AD [86]. Gross examination of the brain in AD
shows a variable degree of cortical atrophy with narrowed
gyri and widened sulci most apparent in the frontal, parietal,
and temporal lobes. Microscopically, the features include
neurofibrillary tangles, neurite (senile) plaques, the central
core of which is amyloid-bpeptide (Ab) derived from the
transmembrane APP, amyloid angiopathy, granulovacuolar
degeneration, and Hirano bodies. Importantly, all of these
changes are present in the brains of nondemented older indi-
viduals but to a much lesser extent [87, 88].
Recent studies have suggested that neuronal death in AD
could arise from dysfunction of the ER. Proteins in the ER
require an efficient system of molecular chaperones whose
role is to assure their proper folding and to prevent accumu-
lation of unfolded proteins. The response of cells to the
accumulation of unfolded proteins in the ER is termed
UPR. UPR is a functional mechanism by which cells
attempt to protect themselves against ER stress, resulting
from the accumulation of the unfolded/misfolded proteins.
Inhibition of protein glycosylation, perturbation of calcium
homeostasis, and reduction of disulfide bonds provoke
accumulation of unfolded protein in the ER, and are called
ER stress. Normal cells respond to ER stress by increasing
transcription of genes encoding ER-resident chaperones
such as GRP78/BiP, to facilitate protein folding or by sup-
pressing the mRNA translation to synthesize proteins.
These systems are termed the UPR. Familial AD-linked
PSEN1 (PS1) mutation downregulates the UPR and leads to
vulnerability to ER stress [89]. Given that AD involves
accumulation of aggregates of two different proteins, the
potential involvement of the UPR and ER dysfunction has
been suggested to lead to cell death. In actively dividing
cells, activation of the UPR is accompanied by decreased
cell cycle protein expression and an arrest in the G1 phase
of the cell cycle [90]. It has been shown that amyloidogenic
proteins can give rise to amyloid fibrils in vitro when a seg-
ment of one of its b-sheets undergoes a conformational
change, exposing an Hsp70 binding site. While normal pro-
teins are rapidly oxidized and subsequently secreted,
mutated proteins remain in the reduced state. Most of these
protein molecules are dislocated out of the ER into the cyto-
sol, where they are ubiquitinylated and degraded by protea-
somes. A parallel pathway for molecules that are not
degraded is condensation into perinuclear aggresomes that
are surrounded by vimentin- or tubulin-containing inter-
mediate filaments and are dependent upon intact microtu-
bules. Inhibition of proteasome activity shifts the balance
toward aggresome formation. Intracellular aggregation is
decreased and targeting to proteasomes improved by over-
expression of the cytosolic chaperone Hsp70. Importantly,
transduction into the cell of an Hsp70 target peptide,
derived from the mutated protein sequence, also reduces
aggresome formation and increases amyloid degradation.
These results demonstrate that amyloidogenic proteins can
aggregate intracellularly despite the common presentation
of extracellular aggregates, and that a similar molecular
surface mediates both in vitro fibril formation and in vivo
aggregation. Furthermore, rationally designed peptides can
be used to suppress this aggregation and may provide a fea-
sible therapeutic approach [91]. Aggresomes are associated
with many neurodegenerative disorders, including AD, PD,
and polyglutamine disorders such as Huntington's disease.
These inclusions commonly contain ubiquitylated proteins.
The stage at which these proteins are ubiquitylated remains
unclear. A malfunction of the ubiquitinproteasome sys-
tem (UPS) may be associated with their formation. Con-
versely, it may reflect an unsuccessful attempt by the cell to
remove them. The 26S proteasome system is involved in
eliminating ubiquitinated misfolded/unfolded proteins, and
its inhibition results in cellular accumulation of protein
aggregates. Accordingly, proteasome dysfunction in AD
neurons may play a role in the accumulation of misfolded,
potentially cytotoxic proteins and may be induced by
increased intracellular AbetaPP/Abeta. Moreover, a role of
microglia recruited from bone marrow (BM) into the CNS
during the progression of AD has been considered and
emerging evidence suggests that infiltration of BM-derived
monocytic cells into the brain contributes to the develop-
ment of microglial reaction in AD, where APP enhances
BM-derived macrophage-mediated clearance of Abeta
[92].
Several lines of evidence support a fundamental role for
calcium, ROS, and RNS secondary to ER, oxidative and
nitrosative stress, respectively, in the pathogenesis of AD
[34, 87, 93]. In particular, neurofibrillary Abhas been
shown to generate both superoxide anion and a-carbon-cen-
tered radicals; in addition the upregulation of iNOS
increases the formation of NO and RNS [94]. Finally, Ab
increases intracellular calcium levels via both the ER rya-
nodine and inositol 1,4,5-trisphosphate receptors thus
inducing ER stress which culminates with the apoptotic
death of neuronal cells [95, 96]. All these prooxidant path-
ways activated by Abcontribute to the massive destruction
1066
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2008, 52, 1062 1073
of brain cells in AD. As mentioned above, antioxidant
enzymes such as HO-1 and TrxR along with the chaperone
Hsp70, are well known intracellular antioxidant systems
which contribute to the “UPR” and guarantee an important
cytoprotective effect against free radical-mediated injury in
AD.
The first evidence of a protective role of curcumin in the
onset of AD derived from epidemiological studies. Ganguli
et al. [97] demonstrated that Indian population, who have a
curcumin-enriched diet, has a reduced prevalence of AD
compared to United States. Following this initial observa-
tion, many basic studies were conducted and the neuropro-
tective role of curcumin was corroborated. In vitro studies
have shown that curcumin protects neuron-like PC12 cells
from b-amyloid toxicity and, interestingly, the polyphenol
displayed a neuroprotective effect greater than a well known
antioxidant such as a-tocopherol [98]. By using an Alz-
heimer transgenic APPSw mouse model (Tg2576), Lim et
al. [99] have shown that dietary curcumin suppressed
inflammation and oxidative damage in the brain of these
mice. More recently, Garcia-Alloza et al. [14] in transgenic
APPswe/PS1dE9 mice demonstrated that curcumin, given
intravenously for 7 days, crosses the blood brain barrier,
binds to b-amyloid deposits in the brain and accelerates
their rate of clearance. These latter results are in good
agreement with previous findings which demonstrated that
curcumin disaggregates and inhibits b-amyloid aggregation
at submicromolar concentrations in vitro, and more impor-
tantly, reduces amyloid levels and plaque burden in vivo in
Tg2576 mice [100, 101]. Curcumin has an important
impact on ER stress biology, as its apoptosis-induced effect
is associated with its ability to cause ER stress. Removing
two double bonds in curcumin, which was speculated to
form Michael adducts with thiols in secretory proteins,
resulted in a loss of the ability of curcumin to induce apop-
tosis as well as ER stress. Inhibition of caspase-4 activity by
z-LEVD-FMK, blockage of survival molecules such as
CAAT/enhancer binding protein homologous protein
(CHOP) expression by small interfering RNA, and treat-
ment with salubrinal, an ER inhibitor, significantly reduced
curcumin-induced apoptosis [102]. In addition to this,
impairment of UPS has been demonstrated to mediate cur-
cumin proapoptotic effects. Curcumin disrupts UPS func-
tion by directly inhibiting the enzyme activity of the protea-
some's 20S core catalytic component. Like other protea-
some inhibitors, curcumin exposure induces neurite out-
growth and the stress response, as evident from the induc-
tion of various cytosolic and ER chaperones as well as
induction of transcription factor CHOP/GADD153. The
direct inhibition of proteasome activity also causes an
increase in half-life of IkBathat ultimately leads to the
downregulation of NF-kappaB activation. These results
suggest that curcumin-induced proteasomal malfunction
might be linked with both antiproliferative and anti-inflam-
matory activities [103].
PD, whose cardinal features include tremor, slowness of
movement, stiffness, and poor balance, is attributed to a
profound deficit in dopamine that follows the loss of dopa-
minergic neurons in the substantia nigra pars compacta
and dopaminergic nerve terminals in the striatum [34, 104].
Although the mechanisms of PD are still uncertain, a large
amount of experimental evidence implicates oxidative and
nitrosative stress as one of the crucial factors in the patho-
genesis of PD [105, 106]. Considerable insights into the
pathogenesis of PD, indeed, have been achieved by use of
the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyri-
dine (MPTP), which is commonly used to induce an experi-
ment model of PD [105, 107]. Excessive free radical forma-
tion or antioxidant deficiency and the resulting oxidative
stress are all mechanisms involved in MPTP neurotoxicity
[108]. Rajeswari [109] has shown that curcumin protects rat
brain from MPTP-induced neurotoxicity by virtue of its
scavenger activity. On the other hand, curcumin has been
shown to protect PC12 cells from MPP+(the active metabo-
lite of MPTP) by inducing the anti-apoptotic protein bcl-2,
preventing the dissipation of mitochondrial membrane
potential and reducing the intracellular iNOS levels [3].
The importance of mitochondria in the neuroprotective
effect of curcumin has been also stressed by Mythri et al.
[110] who demonstrated that curcumin prevents the forma-
tion of peroxynitrite which is responsible for the complex I
damage which is a common feature in PD.
Transient forebrain ischemia is a common cause of
stroke and occurs in people suffering from cardiovascular
diseases [111]. As a consequence of ischemia and the fol-
lowing reperfusion, a cascade of events such as increased
calcium release, the overexpression of cycloxygenase-2
(COX-2) and iNOS both of which are important free-radical
generators and trigger neuronal cell death in selected brain
areas including the hippocampal cornu ammonis 1 (CA1)
[35, 38, 111, 112]. Curcumin exerted a neuroprotective
effect in rats who underwent ischemia/reperfusion injury
and this effect has been related to the direct scavenger effect
of curcumin as well as to a curcumin-induced interference
with the apoptotic machinery, increase in antioxidant mole-
cules (reduced glutathione (GSH)) and enzymes (catalase,
superoxide dismutase) [111, 113, 114].
The mechanism(s) underlying these neuroprotective
effects of curcumin are still debated. Although it is clear
that the free-radical scavenging activity of curcumin plays a
main role in its antioxidant activity, new lines of evidence
propose that curcumin is neuroprotective because of its
ability to induce vitagenes (Figs. 2 and 3). As mentioned
above, curcumin induced HO-1 in rat neurons and astro-
cytes and, consequently the heme-degrading activity of
HO-1 increased [45]. Keeping in mind that through the HO
activity heme is degraded into BV which is then reduced
into BR by biliverdin reductase (BVR) and that BR is a very
efficient scavenger of ROS, NO, and RNS [38, 39, 115
117], it is possible to conclude that the strong antioxidant
1067
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
V. Calabrese et al. Mol. Nutr. Food Res. 2008, 52, 1062 –1073
and anti-inflammatory activities of curcumin could be
mediated by the induction of HO-1.
5 Curcumin and diabetes
Compelling evidence has been provided that both insulin
dependent and noninsulin dependent diabetic patients are
under conditions of oxidative stress and that the complica-
tions of diabetes mellitus could be partially mediated by
oxidative stress [118, 119]. Several mechanisms seem to be
involved in the genesis of oxidative stress in diabetic
patients, including glucose autoxidation, protein glycation,
as well as the formation of advanced-glycation end-prod-
ucts [119, 120].
Recent findings have shown that curcumin can reduce
the degree of systemic oxidative stress as well as retinal
inflammation and diabetic nephropathy in streptozocin-
treated rats, an animal model for diabetes [121 123]. The
protective effect of curcumin and THC in diabetes and its
complications seems to be due to the inhibition of proin-
flammatory cytokines (IL-1b) and growth factors (VEGF),
to the blockade of the NFkB signaling and the increasing
activity of chaperone molecules [122, 124]. Remarkably,
the cytoprotective effect of curcumin in diabetic complica-
tions could be related to the interaction of this spice with
the vitagene system. In fact, type 2 diabetic patients with or
without nephropathy have an increased level of both HO-1
and Hsp70 in lymphocytes and this is considered as an
attempt of the immune system to react to oxidative insults
[125]. The possibility that curcumin may further upregulate
HO-1 and Hsp70 in diabetic patients thus contributing to
counteract prooxidant conditions allows to hypothesize a
potential role of curcumin in the prevention of diabetes and
its complications.
6 Curcumin and other oxidative stress-
related conditions
Oxidative stress plays an important role in the pathogenesis
of lung and liver diseases, both of which recognize oxida-
tive stress as a main pathogenetic factor.
The pharmacological therapy of both asthma and chronic
obstructive pulmonary disease (COPD) is based on the
administration of corticosteroids. Unfortunately, in some
cases, these drugs are ineffective and this is often due to the
development of steroid resistance. Histone deacetylase is
involved in the mechanism of action of corticosteroid and
its activity is often reduced in case of steroid insensitivity
[126]. Curcumin has been shown to restore histone deacety-
lase activity and therefore its use can be hypothesized in the
treatment of lung disease unresponsive to corticosteroids
[126]. However, the concomitant administration of curcu-
min with systemic corticosteroids should be discouraged
because curcumin may inhibit cytochrome P450 and UDP-
glucuronosyltransferases (see above), two enzymatic routes
through which corticosteroids are metabolized.
Curcumin reduces both thioacetamide- and endotoxin-
induced liver dysfunction in rodents and this effect is attrib-
uted to the inhibition of the expression of proinflammatory
cytokines (tumor necrosis factor-aand IL-1b), transcription
factors (NFkB), and enzymes (iNOS) [127 129].
The long term use of some drugs is associated with the
development of organ toxicity often due to increased oxida-
tive stress. Curcumin and THC reduced the kidney toxicity
of chloroquine, gentamicin, and cyclosporin A in the rat [2,
130, 131]. Furthermore, curcumin reduced the indometha-
cin-induced intestinal damage in the rat and the ritonavir-
related vascular dysfunction in porcine coronary arteries
[132, 133]. All these reports agree that the protective effects
of curcumin were due to the well-known ability of this com-
pound to increase both the enzymatic and nonenzymatic
intracellular antioxidant molecules.
Recent evidence highlighted the potential use of curcu-
min in the treatment of cancer [134]. In animal models, cur-
cumin demonstrated an anticancer activity against skin,
colon, lung, and gastrointestinal tumors [134]. Notably, a
phase II clinical trial on patients affected by advanced pan-
creatic cancer and treated with 8 g of curcumin per os for
2 months has shown a biologic activity of this spice in the
therapy of pancreatic cancer [134].
1068
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Figure 3. Curcumin and cellular stress response mediated by
vitagenes. Under conditions of oxidative/nitrosative stress,
there is an increased formation of ROS or RNS. They play a
key role in the pathogenesis of free radical-induced diseases,
such as neurodegenerative disorders. Vitagenes (HO-1 and
Hsp70) contribute to counteract the ROS/RNS-mediated neu-
rotoxic insult, thus initiating a neuroprotective response. In
addition, curcumin by inducing ER stress, disrupting UPS and
downregulating NF-kappaB activation exerts antiproliferative
and anti-inflammatory activities resulting in cytoprotection.
Straight arrows, stimulation; dashed arrows, inhibition.
Mol. Nutr. Food Res. 2008, 52, 1062 1073
7 Conclusions and perspectives
As highlighted in this review, the cytoprotective role of cur-
cumin in several experimental systems is well established.
Commercial grade curcumin contains 10 20% curcumi-
noids, desmethoxycurcumin, and bisdesmethoxycurcumin
and they are as effective as purecurcumin. Based on a num-
ber of clinical studies in carcinogenesis, a daily oral dose of
3.6 g curcumin has been efficacious for colorectal cancer
and advocates its advancement into Phase II clinical stud-
ies. In addition to the anticancer effects, curcumin has been
effective against a variety of disease conditions in both in
vitro and in vivo preclinical studies [135]. Interestingly,
nanodelivery biotechnology, using primarily composed
phospholipids coated with prostate membrane specific anti-
gen specific antibodies, has been developed to enhance tar-
geted delivery of curcumin as an anticancer agent showing
that treatment of human prostate cancer cell lines with lip-
osomal curcumin resulted in at least 70 80% inhibition of
cellular proliferation at ten-fold lower doses compared to
free curcumin [136].
Although curcumin has been described in Ayurveda, as a
treatment for inflammatory diseases and is referred by
different names in different cultures, extensive research
over the last half century has revealed several important
functions, as it binds to a variety of proteins and inhibits the
activity of various kinases. By modulating the activation of
various transcription factors, curcumin regulates the
expression of inflammatory enzymes, cytokines, adhesion
molecules, and cell survival proteins. Curcumin also down-
regulates cyclin D1, cyclin E, and MDM2; and upregulates
p21, p27, and p53. Various preclinical cell culture and ani-
mal studies suggest that curcumin has potential as an anti-
proliferative, anti-invasive, and anti-angiogenic agent; as a
mediator of chemoresistance and radioresistance; as a che-
mopreventive agent; and as a therapeutic agent in wound
healing, diabetes, AD, PD, cardiovascular disease, pulmo-
nary disease, and arthritis. Pilot phase I clinical trials have
shown curcumin to be safe even when consumed at a daily
dose of 12 g for 3 months. Other clinical trials suggest a
potential therapeutic role for curcumin in diseases such as
familial adenomatous polyposis, inflammatory bowel dis-
ease, ulcerative colitis, colon cancer, pancreatic cancer,
hypercholesteremia, atherosclerosis, pancreatitis, psoriasis,
chronic anterior uveitis, and arthritis. Thus, curcumin is
emerging into the clinic and may prove to be “Curecumin”
[137]. However, the potential use of dietary or supplemental
curcumin in the treatment of human pathologies remains to
be refined. Some concern derive from the pharmacokinetics
of curcumin and in particular its poor bioavailability and
metabolic fate [8, 138]. Also, no conclusive data are avail-
able regarding the concentrations of curcumin in the nerv-
ous system as well as the molecular targets of nanomolar
concentrations of curcumin, which are most likely the con-
centrations attained in vivo after regular dietary intake. In
this regard, the possible effects on vitagenes [34, 139] and
related signal transduction pathways which ultimately can
provide cytoprotective and chemopreventive effects open
new targeted strategies for its clinical use. Once better phar-
macodynamics will be achieved then the inhibitory effects
on cytochrome P450 enzymes, GST and UDP-glucurono-
syltransferases elicited by curcumin should be taken into
consideration because many drugs are metabolised by these
enzymes in the liver and gastrointestinal tract and therefore
their inhibition could increase the plasma concentrations
potentiating toxic effects. Keeping all this in mind, it is pos-
sible to conclude that clinical research on curcumin and its
potential use in human diseases needs to be expanded and
therapeutic use of curcumin in several human oxidant stress
pathologies has to be considered a promising strategy.
The authors have declared no conflict of interest.
8 References
[1] Rahman, I., Biswas, S. K., Kirkham, P. A., Regulation of
inflammation and redox signaling by dietary polyphenols,
Biochem. Pharmacol. 2006, 72, 1439 –1452.
[2] Tirkey, N., Kaur, G., Vij, G., Chopra, K., Curcumin, a diferu-
loylmethane, attenuates cyclosporine-induced renal dysfunc-
tion and oxidative stress in rat kidneys, BMC Pharmacol.
2005, 5, 15.
[3] Chen, J., Tang, X. Q., Zhi, J. L., Cui, Y., et al., Curcumin pro-
tects PC12 cells against 1-methyl-4-phenylpyridinium ion-
induced apoptosis by bcl-2-mitochondria-ROS-iNOS path-
way, Apoptosis 2006, 11, 943– 953.
[4] Divya, C. S., Pillai, M. R., Antitumor action of curcumin in
human papillomavirus associated cells involves downregula-
tion of viral oncogenes, prevention of NFkB and AP-1 trans-
location, and modulation of apoptosis, Mol. Carcinog. 2006,
45, 320 –332.
[5] Perez-Arriaga, L., Mendoza-Magana, M. L., Cortes-Zarate,
R., Corona-Rivera, A., et al., Cytotoxic effect of curcumin on
Giardia lamblia trophozoites, Acta Trop. 2006, 98, 152– 161.
[6] Ramsewak, R. S., DeWitt, D. L., Nair, M. G., Cytotoxicity,
antioxidant and anti-inflammatory activities of curcumins I-
III from Curcuma longa, Phytomedicine 2000, 7, 303 –308.
[7] Yang, F., Lim, G. P., Begum, A. N., Ubeda, O. J., et al., Curcu-
min inhibits formation of amyloid beta oligomers and fibrils,
binds plaques, and reduces amyloid in vivo, J. Biol. Chem.
2005, 280, 5892 –5901.
[8] Sharma, R. A., Gescher, A. J., Steward, W. P., Curcumin: The
story so far, Eur. J. Cancer 2005, 41, 1955 –1968.
[9] Ravindranath, V., Chandrasekhara, N., Metabolism of curcu-
min studies with [3H]curcumin, Toxicology 1981– 1982,
22, 337 –344.
[10] Ravindranath, V., Chandrasekhara, N., Absorption and tissue
distribution of curcumin in rats, Toxicology 1980, 16, 259
265.
[11] Maiti, K., Mukherjee, K., Gantait, A., Saha, B. P., Mukherjee,
P. K., Curcumin-phospholipid complex: Preparation, thera-
peutic evaluation and pharmacokinetic study in rats, Int. J.
Pharm. 2007, 330, 155 –163.
1069
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
V. Calabrese et al. Mol. Nutr. Food Res. 2008, 52, 1062 –1073
[12] Shoba, G., Joy, D., Joseph, T., Majeed, M., et al., Influence of
piperine on the pharmacokinetics of curcumin in animals and
human volunteers, Planta Med. 1998, 64, 353 –356.
[13] Cheng, A. L., Hsu, C. H., Lin, J. K., Hsu, M. M., et al., Phase
I clinical trial of curcumin, a chemopreventive agent, in
patients with high-risk or premalignant lesions, Anticancer
Res. 2001, 21, 2895 –2900.
[14] Garcia-Alloza, M., Borrelli, L. A., Rozkalne, A., Hyman, B.
T., Bacskai, B. J., Curcumin labels amyloid pathology in vivo,
disrupts existing plaques, and partially restores distorted neu-
rites in an Alzheimer mouse model, J. Neurochem. 2007, 102,
1095 1104.
[15] Somparn, P., Phisalaphong, C., Nakornchai, S., Unchern, S.,
Morales, N. P., Comparative antioxidant activities of curcu-
min and its demethoxy and hydrogenated derivatives, Biol.
Pharm. Bull. 2007, 30, 74 –78.
[16] Ireson, C. R., Jones, D. J., Orr, S., Coughtrie, M. W., et al.,
Metabolism of the cancer chemopreventive agent curcumin
in human and rat intestine, Cancer Epidemiol. Biomarkers
Prev. 2002, 11, 105 111.
[17] Lin, J. K., Pan, M. H., Lin-Shiau, S. Y., Recent studies on the
biofunctions and biotransformations of curcumin, Biofactors
2000, 13, 153 –158.
[18] Hayeshi, R., Mutingwende, I., Mavengere, W., Masiyanise,
V., Mukanganyama, S., The inhibition of human glutathione
S-transferases activity by plant polyphenolic compounds ella-
gic acid and curcumin, Food Chem. Toxicol. 2007, 45, 286–
295.
[19] Thapliyal, R., Maru, G. B., Inhibition of cytochrome P450
isozymes by curcumins in vitro and in vivo, Food Chem. Tox-
icol. 2001, 39, 541 –547.
[20] Basu, N. K., Ciotti, M., Hwang, M. S., Kole, L., et al., Differ-
ential and special properties of the major human UGT1-
encoded gastrointestinal UDP-glucuronosyltransferases
enhance potential to control chemical uptake, J. Biol. Chem.
2004, 279, 1429 –1441.
[21] Oetari, S., Sudibyo, M., Commandeur, J. N., Samhoedi, R.,
Vermeulen, N. P., Effects of curcumin on cytochrome P450
and glutathione S-transferase activities in rat liver, Biochem.
Pharmacol. 1996, 51, 39 –45.
[22] Sharma, R. A., Euden, S. A., Platton, S. L., Cooke, D. N., et
al., Phase I clinical trial of oral curcumin: Biomarkers of sys-
temic activity and compliance, Clin. Cancer Res. 2004, 10,
6847 6854.
[23] Deodhar, S. D., Sethi, R., Srimal, R. C., Preliminary study on
antirheumatic activity of curcumin (diferuloyl methane),
Indian J. Med. Res. 1980, 71, 632– 634.
[24] Sugiyama, Y., Kawakishi, S., Osawa, T., Involvement of the
beta-diketone moiety in the antioxidative mechanism of tetra-
hydrocurcumin, Biochem. Pharmacol. 1996, 52, 519 525.
[25] Osawa, T., Sugiyama, Y., Inayoshi, M., Kawakishi, S., Anti-
oxidative activity of tetrahydrocurcuminoids. Biosci. Bio-
technol. Biochem. 1995, 59, 1609 1612.
[26] Chen, W. F., Deng, S. L., Zhou, B., Yang, L., Liu, Z. L., Cur-
cumin and its analogues as potent inhibitors of low density
lipoprotein oxidation: H-atom abstraction from the phenolic
groups and possible involvement of the 4-hydroxy-3-methox-
yphenyl groups, Free Radical Biol. Med. 2006, 40, 526 –535.
[27] Priyadarsini, K. I., Maity, D. K., Naik, G. H., Kumar, M. S., et
al., Role of phenolic O-H and methylene hydrogen on the free
radical reactions and antioxidant activity of curcumin, Free
Radic. Biol. Med. 2003, 35, 475 –484.
[28] Reyes-Gordillo, K., Segovia, J., Shibayama, M., Vergara, P.,
et al., Curcumin protects against acute liver damage in the rat
by inhibiting NF-kappaB, proinflammatory cytokines pro-
duction and oxidative stress, Biochim. Biophys. Acta 2007,
1770, 989 –996.
[29] Venkataranganna, M. V., Rafiq, M., Gopumadhavan, S., Peer,
G., et al., NCB-02 (standardized Curcumin preparation) pro-
tects dinitrochlorobenzene-induced colitis through down-reg-
ulation of NFkappa-B and iNOS, World J. Gastroenterol.
2007, 13, 1103 –1107.
[30] Shishodia, S., Potdar, P., Gairola, C. G., Aggarwal, B. B., Cur-
cumin (diferuloylmethane) down-regulates cigarette smoke-
induced NF-kappaB activation through inhibition of Ikappa-
Balpha kinase in human lung epithelial cells: Correlation
with suppression of COX-2, MMP-9 and cyclin D1, Carcino-
genesis 2003, 24, 1269 –1279.
[31] Pan, M. H., Lin-Shiau, S. Y., Lin, J. K., Comparative studies
on the suppression of nitric oxide synthase by curcumin and
its hydrogenated metabolites through down-regulation of
IkappaB kinase and NFkappaB activation in macrophages,
Biochem. Pharmacol. 2000, 60, 1665 –1676.
[32] Bhattacharyya, S., Mandal, D., Sen, G. S., Pal, S., et al.,
Tumor-induced oxidative stress perturbs nuclear factor-kap-
paB activity-augmenting tumor necrosis factor-alpha-medi-
ated T-cell death: Protection by curcumin, Cancer Res. 2007,
67, 362 –370.
[33] Choi, H., Chun, Y. S., Kim, S. W., Kim, M. S., Park, J. W.,
Curcumin inhibits hypoxia-inducible factor-1 by degrading
aryl hydrocarbon receptor nuclear translocator: A mechanism
of tumor growth inhibition, Mol. Pharmacol. 2006, 70,
1664 1671.
[34] Calabrese, V., Guagliano, E., Sapienza, M., Panebianco, M.,
et al., Redox regulation of cellular stress response in aging
and neurodegenerative disorders: Role of vitagenes, Neuro-
chem. Res. 2007, 32, 757 –773.
[35] Mancuso, C., Scapagnini, G., Curr, D., Giuffrida Stella, A.
M., et al., Mitochondrial dysfunction, free radical generation
and cellular stress response in neurodegenerative disorders,
Front. Biosci. 2007, 12, 1107 –1123.
[36] Calabrese, V., Boyd-Kimball, D., Scapagnini, G., Butterfield,
D. A., Nitric oxide and cellular stress response in brain aging
and neurodegenerative disorders: The role of vitagenes, In
Vivo 2004, 18, 245– 267.
[37] Calabrese, V., Butterfield, D. A., Scapagnini, G., Stella, A.
M., Maines, M. D., Redox regulation of heat shock protein
expression by signaling involving nitric oxide and carbon
monoxide: Relevance to brain aging, neurodegenerative dis-
orders, and longevity, Antioxid. Redox Signal. 2006, 8, 444
477.
[38] Mancuso, C., Pani, G., Calabrese, V., Bilirubin: An endoge-
nous scavenger of nitric oxide and reactive nitrogen species,
Redox Rep. 2006, 11, 207 213.
[39] Mancuso, C., Heme oxygenase and its products in the nerv-
ous system, Antioxid. Redox Signal. 2004, 6, 878 –887.
[40] Stocker, R., Antioxidant activities of bile pigments, Antioxid.
Redox Signal. 2004, 6, 841 849.
[41] Abuarqoub, H., Green, C. J., Foresti, R., Motterlini, R., Cur-
cumin reduces cold storage-induced damage in human car-
diac myoblasts, Exp. Mol. Med. 2007, 39, 139– 148.
[42] Jeong, G. S., Oh, G. S., Pae, H. O., Jeong, S. O., et al., Com-
parative effects of curcuminoids on endothelial heme oxy-
genase-1 expression: Ortho-methoxy groups are essential to
enhance heme oxygenase activity and protection, Exp. Mol.
Med. 2006, 38, 393 –400.
1070
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2008, 52, 1062 1073
[43] McNally, S. J., Harrison, E. M., Ross, J. A., Garden, O. J.,
Wigmore, S. J., Curcumin induces heme oxygenase-1 in hep-
atocytes and is protective in simulated cold preservation and
warm reperfusion injury, Transplantation 2006, 81, 623–
626.
[44] Rushworth, S. A., Ogborne, R. M., Charalambos, C. A.,
O'Connell, M. A., Role of protein kinase C delta in curcumin-
induced antioxidant response element-mediated gene expres-
sion in human monocytes, Biochem. Biophys. Res. Commun.
2006, 341, 1007 –1016.
[45] Scapagnini, G., Colombrita, C., Amadio, M., D'Agata, V., et
al., Curcumin activates defensive genes and protects neurons
against oxidative stress, Antioxid. Redox Signal. 2006, 8,
395 403.
[46] Balogun, E., Foresti, R., Green, C. J., Motterlini, R., Changes
in temperature modulate heme oxygenase-1 induction by cur-
cumin in renal epithelial cells, Biochem. Biophys. Res. Com-
mun. 2003, 308, 950 –955.
[47] McNally, S. J., Harrison, E. M., Ross, J. A., Garden, O. J.,
Wigmore, S. J., Curcumin induces heme oxygenase 1 through
generation of reactive oxygen species, p38 activation and
phosphatase inhibition, Int. J. Mol. Med. 2007, 19, 165– 172.
[48] Andreadi, C. K., Howells, L. M., Atherfold, P. A., Manson,
M. M., Involvement of Nrf2, p38, B-Raf, and nuclear factor-
kappaB, but not phosphatidylinositol 3-kinase, in induction
of hemeoxygenase-1 by dietary polyphenols, Mol. Pharma-
col. 2006, 69, 1033 –1040.
[49] Rashmi, R., Santhosh Kumar, T. R., Karunagaran, D., Human
colon cancer cells differ in their sensitivity to curcumin-
induced apoptosis and heat shock protects them by inhibiting
the release of apoptosis-inducing factor and caspases, FEBS
Lett. 2003, 538, 19 –24.
[50] Sood, A., Mathew, R., Trachtman, H., Cytoprotective effect
of curcumin in human proximal tubule epithelial cells
exposed to shiga toxin, Biochem. Biophys. Res. Commun.
2001, 283, 36 –41.
[51] Chen, Y. C., Tsai, S. H., Shen, S. C., Lin, J. K., Lee, W. R.,
Alternative activation of extracellular signal-regulated pro-
tein kinases in curcumin and arsenite-induced HSP70 gene
expression in human colorectal carcinoma cells, Eur. J. Cell.
Biol. 2001, 80, 213 –221.
[52] Chen, Y. C., Kuo, T. C., Lin-Shiau, S. Y., Lin, J. K., Induction
of HSP70 gene expression by modulation of Ca(+2) ion and
cellular p53 protein by curcumin in colorectal carcinoma
cells, Mol. Carcinog. 1996, 17, 224– 234.
[53] Kato, K., Ito, H., Kamei, K., Iwamoto, I., Stimulation of the
stress-induced expression of stress proteins by curcumin in
cultured cells and in rat tissues in vivo, Cell Stress Chaper-
ones 1998, 3, 152 –160.
[54] Fang, J., Lu, J., Holmgren, A., Thioredoxin reductase is irre-
versibly modified by curcumin: A novel molecular mecha-
nism for its anticancer activity, J. Biol. Chem. 2005, 280,
25284 25290.
[55] Talalay, P., Chemoprotection against cancer by induction of
phase 2 enzymes, Biofactors 2000, 12, 5 –11.
[56] Prestera, T., Zhang, Y., Spencer, S. R., Wilczak, C. A., Tala-
lay, P., The electrophile counterattack response: Protection
against neoplasia and toxicity, Adv. Enzyme Regul. 1993, 33,
281 296.
[57] Talalay, P., Mechanisms of induction of enzymes that protect
against chemical carcinogenesis, Adv. Enzyme Regul. 1989,
28, 237 –250.
[58] Prestera, T., Talalay, P., Alam, J., Ahn, Y. I., et al., Parallel
induction of heme oxygenase-1 and chemoprotective phase 2
enzymes by electrophiles and antioxidants: Regulation by
upstream antioxidant-responsive elements (ARE), Mol. Med.
1995, 1, 827 –837.
[59] Wild, A. C., Mulcahy, R. T., Regulation of gamma-glutamyl-
cysteine synthetase subunit gene expression: Insights into
transcriptional control of antioxidant defenses, Free Radic.
Res. 2000, 32, 281 –301.
[60] Nguyen, T., Scherratt, P. J., Pickett, C. B., Regulatory mecha-
nisms controlling gene expression mediated by the antioxi-
dant response element, Ann. Rev. Pharmacol. Toxicol. 2003,
43, 233 –260.
[61] Nioi, P., McMahon, M., Itoh, K., Yamamoto, M., Hayes, J. D.,
Identification of a novel Nrf2-regulated antioxidant response
element (ARE) in the mouse NAD(P)H:quinone oxidoreduc-
tase 1 gene: Reassessment of the ARE consensus sequence,
Biochem. J. 2003, 374, 337– 348.
[62] Moi, P., Chan, K., Asunis, I., Cao, A., Kan, Y. W., Isolation of
NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine
zipper transcriptional activator that binds to the tandem NF-
E2/AP1 repeat of the beta-globin locus control region, Proc.
Natl. Acad. Sci. USA 1994, 91, 9926 –9930.
[63] Itoh, K., Chiba, T., Takahashi, S., Ishii, T., et al., An Nrf2/
small Maf heterodimer mediates the induction of phase II
detoxifying enzyme genes through antioxidant response ele-
ments, Biochem. Biophys. Res. Commun. 1997, 236, 313
322.
[64] Motohashi, H., O
,
Connor, T., Katsuoka, F., Engel, J. D.,
Yamamoto, M., Integration and diversity of the regulatory
network composed of Maf and CNC families of transcription
factors, Gene 2002, 294, 1 –12.
[65] Katsuoka, F., Motohashi, H., Ishii, T., Aburatani, H., et al.,
Genetic evidence that small maf proteins are essential for the
activation of antioxidant response element-dependent genes,
Mol. Cell. Biol. 2005, 25, 8044 –8051.
[66] Motohashi, H., Yamamoto, M., Nrf2-Keap1 defines a physio-
logically important stress response mechanism, Trends Mol.
Med. 2004, 10, 549 –557.
[67] Kensler, T. W., Wakabayashi, N., Biswal, S., Cell survival
responses to environmental stresses via the Keap1-Nrf2-ARE
pathway, Annu. Rev. Pharmacol. Toxicol. 2007, 47, 89 –116.
[68] Kobayashi, M., Yamamoto, M., Nrf2-Keap1 regulation of cel-
lular defense mechanisms against electrophiles and reactive
oxygen species, Adv. Enzyme Regul. 2006, 46, 113 –140.
[69] Itoh, K., Wakabayashi, N., Katoh, Y., Ishii, T., et al., Keap1
represses nuclear activation of antioxidant responsive ele-
ments by Nrf2 through binding to the amino-terminal Neh2
domain, Genes Dev. 1999, 13, 76 86.
[70] McMahon, M., Thomas, N., Itoh, K., Yamamoto, M., Hayes,
J. D., Dimerization of substrate adaptors can facilitate cullin-
mediated ubiquitylation of proteins by a “tethering” mecha-
nism: A two-site interaction model for the Nrf2-Keap1 com-
plex, J. Biol. Chem. 2006, 281, 24756 –24768.
[71] Dinkova-Kostova, A. T., Holtzclaw, W. D., Kensler, T. W.,
The role of Keap1 in cellular protective responses, Chem.
Res. Toxicol. 2005, 18, 1779 –1791.
[72] Dinkova-Kostova, A. T., Holtzclaw, W. D., Cole, R. N., Itoh,
K. et al., Direct evidence that sulfhydryl groups of Keap1 are
the sensors regulating induction of phase 2 enzymes that pro-
tect against carcinogens and oxidants, Proc. Natl. Acad. Sci.
USA 2002, 99, 11908 –11913.
1071
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
V. Calabrese et al. Mol. Nutr. Food Res. 2008, 52, 1062 –1073
[73] Wakabayashi, N., Dinkova-Kostova, A. T., Holtzclaw, W. D.,
Kang, M. I., et al., Protection against electrophile and oxidant
stress by induction of the phase 2 response: Fate of cysteines
of the Keap1 sensor modified by inducers, Proc. Natl. Acad.
Sci. USA 2004, 101, 2040 –2045.
[74] Calabrese, V., Scapagnini, G., Catalano, C., Bates, T. E., et
al., Induction of heat shock protein synthesis in human skin
fibroblasts in response to oxidative stress: Regulation by a
natural antioxidant from rosemary extract, Int. J. Tissue
React. 2001, 23, 51 –58.
[75] Zhang, X., Lu, L., Dixon, C., Wilmer, W., et al., Stress protein
activation by the cyclopentenone prostaglandin 15-deoxy-
D12,14-prostaglandin J2 in human mesangial cells, Kidney
Int. 2004, 65, 798 –810.
[76] Levonen, A. L., Landar, A., Ramachandran, A., Ceaser, E.
K., et al., Cellular mechanisms of redox cell signalling: Role
of cysteine modification in controlling antioxidant defences
in response to electrophilic lipid oxidation products, Bio-
chem. J. 2004, 378, 373– 382.
[77] Ahn, S. G., Thiele, D. J., Redox regulation of mammalian
heat shock factor 1 is essential for Hsp gene activation and
protection from stress, Genes Dev. 2003, 17, 516– 528.
[78] Tabner, B. J., Turnbull, S., El-Agnaf, O., Allsop, D., Produc-
tion of reactive oxygen species from aggregating proteins
implicated in Alzheimer's disease, Parkinson's disease and
other neurodegenerative diseases, Curr. Top. Med. Chem.
2001, 1, 507 –517.
[79] Barnham, K. J., Cappai, R., Beyreuther, K., Masters, C. L.,
Hill, A. F., Delineating common molecular mechanisms in
Alzheimer's and prion diseases, Trends Biochem. Sci. 2006,
31, 465 –472.
[80] Hinault, M. P., Ben-Zvi, A., Goloubinoff, P., Chaperones and
proteases: Cellular fold-controlling factors of proteins in neu-
rodegenerative diseases and aging, J. Mol. Neurosci. 2006,
30, 249 –265.
[81] Butterfield, D. A., Amyloid beta-peptide (1– 42)-induced
oxidative stress and neurotoxicity: Implications for neurode-
generation in Alzheimer's disease brain. A review, Free
Radic. Res. 2002, 36, 1307 –1313.
[82] Zhang, K., Kaufman, R. J., The unfolded protein response: A
stress signaling pathway critical for health and disease, Neu-
rology 2006, 66, S102– S109.
[83] Nakamura, T., Lipton, S. A., Molecular mechanisms of nitro-
sative stress-mediated protein misfolding in neurodegenera-
tive diseases, Cell. Mol. Life Sci. 2007, 64, 1609 –1620.
[84] Schroder, M., The unfolded protein response, Mol. Biotech-
nol. 2006, 34, 279 –290.
[85] Schroder, M., Kaufman, R. J., The mammalian unfolded pro-
tein response, Annu. Rev. Biochem. 2005, 74, 739– 789.
[86] Bird, T. D., Genetic aspects of Alzheimer disease, Genet.
Med. 2008, 10, 231 –239.
[87] Calabrese, V., Sultana, R., Scapagnini, G., Guagliano, E., et
al., Nitrosative stress, cellular stress response, and thiol
homeostasis in patients with Alzheimer's disease, Antioxid.
Redox Signal. 2006, 8, 1975 1986.
[88] Katzman, R., Saitoh, T., Advances in Alzheimer's disease,
FASEB J. 1991, 5, 278 286.
[89] Katayama, T., Imaizumi, K., Manabe, T., Hitomi, J., et al.,
Induction of neuronal death by ER stress in Alzheimer's dis-
ease, J. Chem. Neuroanat. 2004, 28, 67 –78.
[90] Hoozemans, J. J., Stieler, J., van Haastert, E. S., Veerhuis, R.,
Rozemuller, A. J. et al., The unfolded protein response affects
neuronal cell cycle protein expression: Implications for Alz-
heimer's disease pathogenesis, Exp. Gerontol. 2006, 41,
380 386.
[91] Dul, J. L., Davis, D. P., Williamson, E. K., Stevens, F. J.,
Argon, Y., Hsp70 and antifibrillogenic peptides promote
degradation and inhibit intracellular aggregation of amyloi-
dogenic light chains, J. Cell. Biol. 2001, 152, 705– 716.
[92] Yamamoto, M., Kiyota, T., Walsh, S. M., Ikezu, T., Kinetic
analysis of aggregated amyloid-beta peptide clearance in
adult bone-marrow-derived macrophages from APP and
CCL2 transgenic mice, J. Neuroimmune Pharmacol. 2007,
2, 213 –221.
[93] Guix, F. X., Uribesalgo, I., Coma, M., Munoz, F. J., The
physiology and pathophysiology of nitric oxide in the brain,
Prog. Neurobiol. 2005, 76, 126 152.
[94] Mancuso, C., Bates, T. E., Butterfield, D. A., Calafato, S., et
al., Natural antioxidants in Alzheimer's disease, Expert
Opin. Investig. Drugs 2007, 12, 1921 1931.
[95] Ferreiro, E., Resende, R., Costa, R., Oliveira, C. R., Pereira,
C. M., An endoplasmic-reticulum-specific apoptotic path-
way is involved in prion and amyloid-beta peptides neuro-
toxicity, Neurobiol. Dis. 2006, 23, 669 678.
[96] Ferreiro, E., Oliveira, C. R., Pereira, C., Involvement of
endoplasmic reticulum Ca2+ release through ryanodine and
inositol 1,4,5-triphosphate receptors in the neurotoxic
effects induced by the amyloid-beta peptide, J. Neurosci.
Res. 2004, 76, 872 –880.
[97] Ganguli, M., Chandra, V., Kamboh, M. I., Johnston, J. M., et
al., Apolipoprotein E polymorphism and Alzheimer disease:
The Indo-US Cross-National Dementia Study, Arch. Neurol.
2000, 57, 824 –830.
[98] Kim, D. S., Park, S. Y., Kim, J. K., Curcuminoids from Cur-
cuma longa L. (Zingiberaceae) that protect PC12 rat pheo-
chromocytoma and normal human umbilical vein endothe-
lial cells from betaA(1 42) insult, Neurosci. Lett. 2001,
303, 57 –61.
[99] Lim, G. P., Chu, T., Yang, F., Beech, W., et al., The curry
spice curcumin reduces oxidative damage and amyloid path-
ology in an Alzheimer transgenic mouse, J. Neurosci. 2001,
21, 8370 –8377.
[100] Ono, K., Hasegawa, K., Naiki, H., Yamada, M., Curcumin
has potent anti-amyloidogenic effects for Alzheimer's beta-
amyloid fibrils in vitro, J. Neurosci. Res. 2004, 75, 742–
750.
[101] Begum, A. N., Jones, M. R., Lim, G. P., Morihara, T., et al.,
Curcumin structure-function, bioavailablity and efficacy in
models of neuroinflammation and Alzheimer's, J. Pharma-
col. Exp. Ther. 2008, 326, 196 208.
[102] Pae, H. O., Jeong, S. O., Jeong, G. S., Kim, K. M., et al., Cur-
cumin induces proapoptotic endoplasmic reticulum stress in
human leukemia HL-60 cells, Biochem. Biophys. Res. Com-
mun. 2007, 353, 1040 –1045.
[103] Dikshit, P., Goswami, A., Mishra, A., Chatterjee, M., Jana,
N. R., Curcumin induces stress response, neurite outgrowth
and prevent NF-kappaB activation by inhibiting the protea-
some function, Neurotox. Res. 2006, 9, 29 37.
[104] Voelker, R., Parkinson disease guidelines aid diagnosis,
management, JAMA 2006, 295, 2126 2128.
[105] Przedborski, S., Ischiropoulos, H., Reactive oxygen and
nitrogen species: Weapons of neuronal destruction in models
of Parkinson's disease, Antioxid. Redox Signal. 2005, 7,
685 693.
[106] Hald, A., Lotharius, J., Oxidative stress and inflammation in
Parkinson's disease: Is there a causal link? Exp. Neurol.
2005, 193, 279 –290.
1072
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2008, 52, 1062 1073
[107] Burns, R. S., Chiueh, C. C., Markey, S. P., Ebert, M. H., et
al., A primate model of parkinsonism: Selective destruction
of dopaminergic neurons in the pars compacta of the sub-
stantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyri-
dine, Proc. Natl. Acad. Sci. USA 1983, 80, 4546 4550.
[108] Chiueh, C. C., Andoh, T., Lai, A. R., Lai, E., Krishna, G.,
Neuroprotective strategies in Parkinson's disease: Protection
against progressive nigral damage induced by free radicals,
Neurotox. Res. 2000, 2, 293– 310.
[109] Rajeswari, A., Curcumin protects mouse brain from oxida-
tive stress caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
pyridine, Eur. Rev. Med. Pharmacol. Sci. 2006, 10, 157
161.
[110] Mythri, R. B., Jagatha, B., Pradhan, N., Andersen, J., Bhar-
ath, M. M., Mitochondrial complex I inhibition in Parkin-
son's disease: How can curcumin protect mitochondria?
Antioxid. Redox Signal. 2007, 9, 399 –408.
[111] Al-Omar, F. A., Nagi, M. N., Abdulgadir, M. M., Al Joni, K.
S., Al-Majed, A. A., Immediate and delayed treatments with
curcumin prevents forebrain ischemia-induced neuronal
damage and oxidative insult in the rat hippocampus, Neuro-
chem. Res. 2006, 31, 611 618.
[112] Mancuso, C., Perluigi, M., Cini, C., De Marco, C., et al.,
Heme oxygenase and cyclooxygenase in the central nervous
system: A functional interplay, J. Neurosci. Res. 2006, 84,
1385 1391.
[113] Wang, Q., Sun, A. Y., Simonyi, A., Jensen, M. D., et al., Neu-
roprotective mechanisms of curcumin against cerebral
ischemia-induced neuronal apoptosis and behavioral defi-
cits, J. Neurosci. Res. 2005, 82, 138 148.
[114] Ghoneim, A. I., Abdel-Naim, A. B., Khalifa, A. E., El-
Denshary, E. S., Protective effects of curcumin against
ischaemia/reperfusion insult in rat forebrain, Pharmacol.
Res. 2002, 46, 273 –279.
[115] Mancuso, C., Bonsignore, A., Capone, C., Di Stasio, E.,
Pani, G., Albumin-bound bilirubin interacts with nitric oxide
by a redox mechanism, Antioxid. Redox Signaling 2006, 8,
487 494.
[116] Mancuso, C., Bonsignore, A., Di Stasio, E., Mordente, A.,
Motterlini, R., Bilirubin and S-nitrosothiols interaction: Evi-
dence for a possible role of bilirubin as a scavenger of nitric
oxide, Biochem. Pharmacol. 2003, 66, 2355 –2363.
[117] Minetti, M., Mallozzi, C., Di Stasi, A. M., Pietraforte, D.,
Bilirubin is an effective antioxidant of peroxynitrite-medi-
ated protein oxidation in human blood plasma, Arch. Bio-
chem. Biophys. 1998, 352, 165 –174.
[118] Ceriello, A., Oxidative stress and diabetes-associated com-
plications, Endocr. Pract. 2006, 12, 60 62.
[119] Dav, G., Falco, A., Patrono, C., Lipid peroxidation in diabe-
tes mellitus, Antioxid. Redox Signaling 2005, 7, 256 –268.
[120] Bonnefont-Rousselot, D., Bastard, J. P., Jaudon, M. C.,
Delattre, J., Consequences of the diabetic status on the oxi-
dant/antioxidant balance, Diabetes Metab. 2000, 26, 163–
176.
[121] Murugan, P., Pari, L., Effect of tetrahydrocurcumin on
plasma antioxidants in streptozotocin-nicotinamide experi-
mental diabetes, J. Basic Clin. Physiol. Pharmacol. 2006,
17, 231 –244.
[122] Kowluru, R. A., Kanwar, M., Effects of curcumin on retinal
oxidative stress and inflammation in diabetes, Nutr. Metab.
(Lond). 2007, 4,8.
[123] Sharma, S., Kulkarni, S. K., Chopra, K., Curcumin, the
active principle of turmeric (Curcuma longa), ameliorates
diabetic nephropathy in rats, Clin. Exp. Pharmacol. Physiol.
2006, 33, 940 –945.
[124] Kumar, P. A., Suryanarayana, P., Reddy, P. Y., Reddy, G. B.,
Modulation of alpha-crystallin chaperone activity in dia-
betic rat lens by curcumin, Mol. Vis. 2005, 11, 561 568.
[125] Calabrese, V., Mancuso, C., Sapienza, M., Guagliano, E., et
al., Oxidative stress and cellular stress response in diabetic
nephropathy, Cell Stress Chaperones 2007, 12, 299– 306.
[126] Marwick, J. A., Ito, K., Adcock, I. M., Kirkham, P. A., Oxi-
dative stress and steroid resistance in asthma and COPD:
Pharmacological manipulation of HDAC-2 as a therapeutic
strategy, Expert Opin. Ther. Targets 2007, 11, 745 –755.
[127] Bruck, R., Ashkenazi, M., Weiss, S., Goldiner, I., et al., Pre-
vention of liver cirrhosis in rats by curcumin, Liver Int.
2007, 27, 373 –383.
[128] Shapiro, H., Ashkenazi, M., Weizman, N., Shahmurov, M.,
et al., Curcumin ameliorates acute thioacetamide-induced
hepatotoxicity, J. Gastroenterol. Hepatol. 2006, 21, 358
366.
[129] Kaur, G., Tirkey, N., Bharrhan, S., Chanana, V., et al., Inhib-
ition of oxidative stress and cytokine activity by curcumin in
amelioration of endotoxin-induced experimental hepatoxic-
ity in rodents, Clin. Exp. Immunol. 2006, 145, 313 –321.
[130] Farombi, E. O., Ekor, M., Curcumin attenuates gentamicin-
induced renal oxidative damage in rats, Food Chem. Toxicol.
2006, 44, 1443 –1448.
[131] Pari, L., Murugan, P., Tetrahydrocurcumin: Effect on chloro-
quine-mediated oxidative damage in rat kidney, Basic Clin.
Pharmacol. Toxicol. 2006, 99, 329– 334.
[132] Sivalingam, N., Hanumantharaya, R., Faith, M., Basivir-
eddy, J. et al., Curcumin reduces indomethacin-induced
damage in the rat small intestine, J. Appl. Toxicol. 2007, 27,
551 560.
[133] Chai, H., Yan, S., Lin, P., Lumsden, A. B., et al., Curcumin
blocks HIV protease inhibitor ritonavir-induced vascular
dysfunction in porcine coronary arteries, J. Am. Coll. Surg.
2005, 200, 820 –830.
[134] Shishodia, S., Chaturvedi, M. M., Aggarwal, B. B., Role of
curcumin in cancer therapy, Curr. Probl. Cancer 2007, 31,
243 305.
[135] Jacob, A., Wu, R., Zhou, M., Wang, P., Mechanism of the
Anti-inflammatory effect of curcumin: PPAR-gamma acti-
vation, PPAR Res. 2007, 2007, 89369.
[136] Thangapazham, R. L., Puri, A., Tele, S., Blumenthal, R.,
Maheshwari, R. K., Evaluation of a nanotechnology-based
carrier for delivery of curcumin in prostate cancer cells, Int.
J. Oncol. 2008, 32, 1119– 1123.
[137] Goel, A., Kunnumakkara, A. B., Aggarwal, B. B., Curcumin
as “Curecumin”: From kitchen to clinic, Biochem. Pharma-
col. 2008, 75, 787 –809.
[138] Garcea, G., Berry, D. P., Jones, D. J., Singh, R., et al., Con-
sumption of the putative chemopreventive agent curcumin
by cancer patients: Assessment of curcumin levels in the col-
orectum and their pharmacodynamic consequences, Cancer
Epidemiol. Biomarkers Prev. 2005, 14, 120 125.
[139] Calabrese, V., Mancuso, C., Calvani, M., Rizzarelli, E., et
al., Nitric oxide in the central nervous system: Neuroprotec-
tion versus neurotoxicity, Nat. Rev. Neurosci. 2007, 8, 766
775.
1073
i2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
... Curcumin has a strong antioxidant effect; it can both directly neutralize free radicals and induce the body's antioxidant enzymes (Agarwal et al., 2010). It has also been reported to activate intracellular anti-oxidative defense mechanisms and eliminate free radicals under both in vitro and in vivo conditions (Calabrese et al., 2008;Wu et al., 2019). However, the effects of curcumin supplementation on buffalo MECs have not been explored, and this is the major focus of the present study. ...
... This temperature (42 • C) has been considered a positive control group to explore whether the buffalo MECs cultured with different doses of curcumin can survive the heat shock and maintain their normal functions. Curcumin, widely used as a major ingredient in Asian cuisine, has been reported to reduce oxidative stress, enhance immune response, and maintain normal cellular functions during stress (Calabrese et al., 2008). Indeed, we have demonstrated in our present study that lower doses of curcumin (up to 20 µM) could alleviate the unfavorable effects of hyperthermia on the viability and proliferation of BuMECs. ...
Article
The epithelial cell is the main basic unit of the udder in which milk synthesis takes place. Curcumin is well known for its antioxidant, anti-apoptotic, and anti-inflammatory properties. The present study was performed to test whether in vitro curcumin supplementation can alleviate the unfavorable impact of hyperthermia on buffalo mammary epithelial cells (BuMECs). The spontaneously immortalized BuMECs were divided into 7 groups (n = 9); 1) unstressed BuMECs (negative control, 37 • C); 2) BuMECs exposed to hyperthermia without curcumin treatment (positive control); 3-7) BuMECs cultured with different concentrations of curcumin (5, 10, 20, 40 and 60 μM), respectively, followed by hyperthermic exposure (42ºC) for 1 h and then returned to 37ºC. Changes in viability (MTT assay), proliferation (BrdU colorimetric immunoassay) and concentrations of antioxidant enzymes , CAT, and SOD (ELISA) of BuMECs were recorded. The gene expression study was performed using qRT-PCR. Lower concentrations of curcumin (5, 10 μM) maintained viability, enhanced proliferation, and content of antioxidant enzymes of heat stressed BuMECs. Curcumin induced thermotolerance and antioxidant status by upregulating the expression of antioxidants genes, anti-apoptotic genes and heat shock proteins in heat stressed BuMECs compared to the positive control group. Besides, curcumin reduced apoptosis and inflammation in BuMECs exposed to hyperthermia by downregulating the expression of genes and transcriptional factors associated with apoptosis and inflammatory immune response. The results reveal the potential roles of curcumin in eliminating the negative impact of hyperthermia on BuMECs by regulating the pathways of apoptosis, inflammation , and oxidative stress.
... Piperine is a hydrophobic polyphenol derived from the black pepper plant. (Figure 1) [10,12]. ...
... Curcumin is a chemical component found in Curcuma longa (turmeric) rhizomes as well as other Curcuma spp, also referred to as diferuloylmethane. Curcumin-mediated inhibition of nuclear Factor-B (NF-kB), the primary device in the inflammatory cascade, was one of the most important discoveries in medicine (26). Curcumin also impacts several signaling pathways linked to cancer (27). ...
Article
Full-text available
Breast cancer is the most common type of cancer and the second most common cause of death. Influential and widely accepted breast cancer prevention strategies are still elusive. Aspirin and other nonsteroidal anti-inflamma-tory drugs, such as ibuprofen, are frequently recommended to treat fever, pain, and inflammation. Studies have shown that these medications also have anticancer effects. A key target of tumor-targeted therapy has been discovered in recent years, and it is the PI3K/AKT/mTOR signalling pathway. In order for tumour cells to proliferate, develop, migrate, and survive, the PI3K/AKT/mTOR signalling system is essential. So, using in silico analysis, we chose to determine whether the NSAID Diclofenac and other phytochemicals have anticancer properties and also act as PI3K/AKT/mTOR inhibitors. We conducted docking analyses for the drugs diclofenac, piperine, naringin, eugenol, d-lim-onene, cinnamaldehyde, and curcumin versus the FDA-approved AKT/PI3K and mTOR inhib-itors alpelisib, rapamycin, and afuresertib. Both bioactivity and ADMET profiling were done for these drugs. We concluded from our in silico analysis that diclofenac, d-limonene, and piper-ine do not contradict any of Lipinski's rules and can be effective AKT/PI3K/mTOR inhibitors.
... Furthermore, curcumin exhibits antioxidant properties, enabling it to reduce oxidative stress. Reactive oxygen species play a role in neuroinflammation and in neurodegenerative processes (87). By neutralizing reactive oxygen species (ROS) and enhancing antioxidant defenses, curcumin may protect neurons from oxidative damage. ...
Article
Full-text available
Uncontrolled, chronic inflammation in the retina can disturb retinal structure and function leading to impaired visual function. For the first time, in a mouse model of chronic neuroinflammation (GFAP-IL6), we investigated the impact of chronic glial activation on the retinal microglia population and structure. In addition, we tested a curcumin Phytosome TM preparation with enhanced bioavailability to investigate the effects of a cytokine-suppressing anti-inflammatory drug on retinal architecture. Curcumin Phytosome TM was fed to 3-month old GFAP-IL6 mice for 4 weeks and compared to their untreated GFAP-IL6 counterparts as well as wild type mice on control diet. Microglial numbers and morphology together with neuronal numbers were characterized using immunohistochemistry and cell reconstruction in the retina, using retinal wholemount and slices. GFAP-IL6 mice showed a significant increase in Iba1-labelled mononuclear phagocytes, including microglia, and displayed altered glial morphology. This resulted in a reduction in cone density and a thinning of the retinal layers compared to wild type mice. Curcumin Phytosome TM treatment contributed to decreased microglial density, significantly decreasing both soma and cell size compared to control diet, as well as preventing the thinning of the retinal layers. This study is the first to characterize the impact of chronic retinal inflammation in the GFAP-IL6 mouse and the therapeutic benefit of enhanced bioavailable curcumin Phytosome TM to significantly reduce microglia density and prevent neuronal loss. These data suggest that curcumin could be used as a complementary therapy alongside traditional treatments to reduce associated retinal inflammation in a variety of retinal diseases. Pé rez-Ferná ndez V, Thananjeyan AL, Ullah F, Münch G, Cameron M and Gyengesi E (2023) The effects of a highly bioavailable curcumin Phytosome TM preparation on the retinal architecture and glial reactivity in the GFAP-IL6 mice.
... 96 cancer is influenced by free radicals and other hazardous byproducts of oxidative stress, and curcumin possesses antioxidant properties that lessen or prevent free radical damage. 97 curcumin inhibits the growth of tumors through a combi- 129 According to some studies, the extract of Polygonum aviculare significantly reduced the growth of HeLa cancer cells. 130,131 The plant extracts can cause cytotoxicity in breast cancer cells by inducing apoptosis. ...
... Curuminoids are treat inflammation in the joints and tendons due to the antioxidant feature of it as a result curuminoids had returned the leukocytes count and erythrocyte sedimentation rate into normal values in the blood so, these results were agreement with Calabrese et al., [26] , Barzegar and moosavimovahedi [25]. However, as the study result considerable increase (p<0.05) in the average of WBCs in the groups of rats treated with Cur + Vit C compare with Cur , VitC and control groups may be according to inflammatory response to treat . ...
Article
The Aim: The goal of this study was to see if curcumin and vitamin C might protect male rats' blood from oxidative stress caused by naproxen using hematological criteria and antioxidant measures. Study Design: The rats were aged 2.5-3 months and contemplated (200-250) gm. The purpose of this study was to see if Cur and Vitamin C might protect male rats against oxidative damage caused by NP, as well as hematological. Place and Duration of Study: From April to October 2019, the experiment was conducted out on 48 male rats weighing between (200-250) gm and aged 2.5-3 months at the animal house of the faculty of science/university of Kufa. Methodology: The 48 male Wister albino rats were separated into six groups at random; Group (1) NP was administered orally to rats at a dose of 40 mg/kg (as positive control). Group (2) cur at a dose of 150 mg/kg was given orally to the rats as a treatment. Group (3) During a 14-day period, rats were given 150 mg/kg of vitamin C by oral administration. Group (4) cur (150 mg/kg) and Vitamin C (150 mg/kg) were given orally to the rats as a treatment. Group (5) The oral administration of NP (40 mg/kg) plus Cur (150 mg/kg) plus Vit C (150 mg/kg) was performed on rats. Group (6) normal saline solution was given to the rats orally during the experiment (as negative control). Results: Finishing the experiments, the findings displayed a considerable decrease (p<0.05) in the average of body weight, hemoglobin concentration (HB) and haematocrit ( HCT) while considerable increase (p<0.05) in the white blood cells(WBC) counts in the animals treated with NP compared to the control and other treated groups , while the results offered considerable increase (p <0.05) in the average of body contemplate , HB, HCT and a considerable decrease (p<0.05) in WBC rate in the animals treated with Cur, VitC, Cur+Vit C, Cur +Vit C+ Np compared the control group . Conclusion: In conclusion, whereas naproxen has a harmful impact on the body by increasing oxidative stress, curcumin and vitamin C operate as powerful antioxidant supplements that reduce malonal di aldehyde (MDA) and boost overall antioxidant capacity by increasing superoxide dismutase (SOD) in the body.
... Meanwhile, the signaling pathways affected by curcuminoids were extended to TP53, MAPKs, Akt, Notch-1, Nrf2, JAK/STAT, Wnt/β-Catenin, AMPK/COX-2 and others [79]. From 2008, these in vitro data started to extend into the clinic through clinical trials [80], which have also been recently reviewed [17]. ...
Article
Full-text available
The number of published studies on curcuminoids in cancer research, including its lead molecule curcumin and synthetic analogs, has been increasing substantially during the past two decades. Insights on the diversity of inhibitory effects they have produced on a multitude of pathways involved in carcinogenesis and tumor progression have been provided. As this wealth of data was obtained in settings of various experimental and clinical data, this review first aimed at presenting a chronology of discoveries and an update on their complex in vivo effects. Secondly, there are many interesting questions linked to their pleiotropic effects. One of them, a growing research topic, relates to their ability to modulate metabolic reprogramming. This review will also cover the use of curcuminoids as chemosensitizing molecules that can be combined with several anticancer drugs to reverse the phenomenon of multidrug resistance. Finally, current investigations in these three complementary research fields raise several important questions that will be put among the prospects for the future research related to the importance of these molecules in cancer research.
Article
Full-text available
Cancer is one of the leading causes of death, despite advancements in health technologies and therapies. While effective, the treatment, chemotherapy can come with huge side effects. This review explores the diverse world of medicinal plants as potential cancer fighters. Numerous plant extracts have shown impressive anti-cancer properties in laboratory settings, offering a natural approach with potentially fewer side effects. However, further research is crucial to evaluate their efficacy and safety in human patients. This review will study the various medicinal plants and the chemical compounds that are promising as anticancer agents and will outline their potential mechanism of action.
Article
Background G protein‐coupled receptor 40 (GPR40) is a potential drug target for Alzheimer's disease (AD), and its agonist GW9508 ameliorates cognitive impairment by intravenous administration. Objectives The present study was conducted to investigate the efficacy of GW9508 administered peripherally on cognitive dysfunction in streptozotocin (STZ)‐induced AD mice. Methods Seventy male ICR mice were randomly divided into seven groups: vehicle sham group, model, Donepezil, GW9508‐L, GW9508‐M, GW9508‐H, and GW1100 + GW9508‐H groups, and administered either vehicle (artificial cerebrospinal fluid [aCSF]) or STZ (3 mg/kg in the vehicle) once a day (9:00 a.m.) by intracerebroventricular injection bilaterally on day 1 and day 3, respectively. After 2 weeks of recovery, all mice were given drug treatment. Behavioral experiments were applied to test the recognition and spatial memory of mice, while molecular biology experiments such as Western blot, ELISA, and Nissl staining were used to detect the corresponding changes of signaling pathways. Results Intraperitoneal administration of GW9508 prevented STZ‐induced cognitive impairment as well as decreased the level of p‐tau and Aβ 1–42 in plasma and brain. GW9508 upregulated the expression of gut‐brain peptides like PYY, CCK, IGF‐1, and GLP‐1 both in blood circulation and brain and downregulated the expression level of autophagy‐related proteins through activating Akt/mTOR signaling pathway. Meanwhile, the treatment effect of GW9508 was reversed by GPR40 antagonist GW1100 significantly. Conclusion Peripheral administration of GW9508 exhibits neuroprotective effects, and it could be a promising therapy for AD. The neuroprotective mechanism of GW9508 was based on promoting gut‐brain peptide secretion, activating Akt/mTOR signal pathway, and regulating neuronal autophagy.
Article
Full-text available
The formation of toxic protein aggregates is a common denominator to many neurodegenerative diseases and aging. Accumulation of toxic, possibly infectious protein aggregates induces a cascade of events, such as excessive inflammation, the production of reactive oxygen species, apoptosis and neuronal loss. A network of highly conserved molecular chaperones and of chaperone-related proteases controls the fold-quality of proteins in the cell. Most molecular chaperones can passively prevent protein aggregation by binding misfolding intermediates. Some molecular chaperones and chaperone-related proteases, such as the proteasome, can also hydrolyse ATP to forcefully convert stable harmful protein aggregates into harmless natively refoldable, or protease-degradable, polypeptides. Molecular chaperones and chaperone-related proteases thus control the delicate balance between natively folded functional proteins and aggregation-prone misfolded proteins, which may form during the lifetime and lead to cell death. Abundant data now point at the molecular chaperones and the proteases as major clearance mechanisms to remove toxic protein aggregates from cells, delaying the onset and the outcome of protein-misfolding diseases. Therapeutic approaches include treatments and drugs that can specifically induce and sustain a strong chaperone and protease activity in cells and tissues prone to toxic protein aggregations.
Chapter
The unfolded protein response is a regulatory mechanism that enhances the expression of proteins involved in the function of the endoplasmic reticulum (ER), including ER chaperones as well as components of ER-associated degradation, when eukaryotic cells increase the production of secretory proteins and the capacity of the ER function is overwhelmed. Without proper functioning of the unfolded protein response, secretory recombinant proteins produced in the ER cannot be correctly folded, are detained in the ER, and evoke ER stress, resulting in apoptotic cell death. Thus, the unfolded protein response is one of the most critical elements for efficient production of recombinant proteins in eukaryotic cells. In this article, the basic information and recent progress in the unfolded protein response of yeast and mammals will be summarized. The mechanisms regulating the capacity of organelles other than the ER, such as the Golgi stress response, lysosome stress response, mitochondrial unfolded protein response, and peroxisomal stress response, will be described. This information will be beneficial to construct ‘suprasecretory cells’, which acquire enormous capacity to produce recombinant secretory proteins indispensable in the biotechnology industry.
Article
γ-Glutamylcysteine synthetase (GCS; also referred to as glutamate-cysteine ligase, GLCL) catalyzes the rate-limiting reaction in glutathione (GSH) biosynthesis. The GCS holoenzyme is composed of a catalytic and regulatory subunit, each encoded by a unique gene. In addition to some conditions which specifically upregulate the catalytic subunit gene, expression of both genes is increased in response to many Phase II enzyme inducers including oxidants, heavy metals, phenolic antioxidants and GSH-conjugating agents. Electrophile Response Elements (EpREs), located in 5′-flanking sequences of both the GCSh and GCSl subunit genes, are hypothesized to at least partially mediate gene induction following xenobiotic exposure. Recent experiments indicate that the bZip transcription factor Nrf2 participates in EpRE-mediated GCS subunit gene activation in combination with other bZip proteins. An AP-1-like binding sequence and an NF-κB site have also been implicated in regulation of the catalytic subunit gene following exposure to certain pro-oxidants. Potential signaling mechanisms mediating GCS gene induction by the diverse families of Phase II enzyme inducers include thiol modification of critical regulatory sensor protein(s) and the generation of the reactive oxygen species. This review summarizes recent progress in defining the molecular mechanisms operative in transcriptional control of the genes encoding the two GCS subunits, identifying areas of agreement and controversy. The mechanisms involved in GCS regulation might also be relevant to the transcriptional control of other components of the antioxidant defense battery.
Article
Curcumin has been shown to induce apoptosis in many cancer cells. However, the molecular mechanism(s) responsible for curcumin-induced apoptosis is not well understood and most probably involves several pathways. In HL-60 cells, curcumin induced apoptosis and endoplasmic reticulum (ER) stress as evidenced by the survival molecules such as phosphorylated protein kinase-like ER-resident kinase, phosphorylated eukaryotic initiation factor-2α, glucose-regulated protein-78, and the apoptotic molecules such as caspase-4 and CAAT/enhancer binding protein homologous protein (CHOP). Inhibition of caspase-4 activity by z-LEVD-FMK, blockage of CHOP expression by small interfering RNA, and treatment with salubrinal, an ER inhibitor, significantly reduced curcumin-induced apoptosis. Removing two double bonds in curcumin, which was speculated to form Michael adducts with thiols in secretory proteins, resulted in a loss of the ability of curcumin to induce apoptosis as well as ER stress. Thus, the present study shows that curcumin-induced apoptosis is associated with its ability to cause ER stress.
Article
Curcumin is a major component of Curcuma species, which is commonly used as a yellow coloring and flavoring agent in foods. Curcumin has shown anti-carcinogenic activity in animals as indicated by its ability to block colon tumor initiation by azoxymethane and skin tumor promotion induced by phorbol ester TPA. Curcumin possesses anti-inflammatory activity and is a potent inhibitor of reactive oxygen-generating enzymes such as lipoxygenase/cyclooxygenase, xanthine dehydrogenase/oxidase and inducible nitric oxide synthase. Curcumin is also a potent inhibitor of protein kinase C, EGF-receptor tyrosine kinase and IκB kinase. Subsequently, curcumin inhibits the activation of NFκB and the expressions of c-jun, c-fos, c-myc and iNOS. It is proposed that curcumin may suppress tumor promotion through blocking signal transduction pathways in the target cells. Curcumin was first biotransformed to dihydrocurcumin and tetrahydrocurcumin and that these compounds subsequently were converted to monoglucuronide conjugates. These results suggest that curcumin-glucuronide, dihydro-curcumin-glucuronide, tetrahydrocurcumin-glucuronide and tetrahydrocurcumin are major metabolites of curcumin in mice.
Article
Curcumin (diferuloyl methane) is the major active yellow pigment of turmeric and curry. Studies in recent years have indicated that curcumin is a potent inhibitor of the initiation and promotion of chemical carcinogen-induced skin carcinogenesis in mice. When COLO205 colorectal carcinoma cells were treated with curcumin (60 μM), the appearance of apoptotic DNA ladders was delayed about 5 h, and G1 arrest was detected. Further analysis of the endonuclease activities in these cells revealed that the activity of Ca+2-dependent endonuclease in COLO205 cells was profoundly inhibited and that the extent of inhibition depended on the degree of calcium depletion. The reduction of p53 gene expression was accompanied by the induction of HSP70 gene expression in the curcumin-treated cells. These findings suggest that curcumin may induce the expression of the HSP70 gene through the initial depletion of intracellular Ca+2, followed by the suppression of p53 gene function in the target cells. © 1996 Wiley-Liss, Inc.
Article
The induction of phase II detoxifying enzymes is an important defense mechanism against intake of xenobiotics. While this group of enzymes is believed to be under the transcriptional control of antioxidant response elements (AREs), this contention is experimentally unconfirmed. Since the ARE resembles the binding sequence of erythroid transcription factor NF-E2, we investigated the possibility that the phase II enzyme genes might be regulated by transcription factors that also bind to the NF-E2 sequence. The expression profiles of a number of transcription factors suggest that an Nrf2/small Maf heterodimer is the most likely candidate to fulfill this rolein vivo.To directly test these questions, we disrupted the murinenrf2 genein vivo.While the expression of phase II enzymes (e.g., glutathione S-transferase and NAD(P)H: quinone oxidoreductase) was markedly induced by a phenolic antioxidantin vivoin both wild type and heterozygous mutant mice, the induction was largely eliminated in the liver and intestine of homozygousnrf2-mutant mice. Nrf2 was found to bind to the ARE with high affinity only as a heterodimer with a small Maf protein, suggesting that Nrf2/small Maf activates gene expression directly through the ARE. These results demonstrate that Nrf2 is essential for the transcriptional induction of phase II enzymes and the presence of a coordinate transcriptional regulatory mechanism for phase II enzyme genes. Thenrf2-deficient mice may prove to be a very useful model for thein vivoanalysis of chemical carcinogenesis and resistance to anti-cancer drugs.
Article
Nitric oxide (NO) plays an important role in inflammation and in the multiple stages of carcinogenesis. In this study, we investigated the inhibitory effects of curcumin and its metabolites, tetrahydrocurcumin, hexahydrocurcumin, and octahydrocurcumin, on the induction of NO synthase (NOS) in RAW 264.7 cells activated with lipopolysaccharide (LPS). Western blotting and northern blotting analyses demonstrated that curcumin strongly reduced 130-kDa protein and 4.5-kb mRNA levels of iNOS in LPS-activated macrophages compared with its metabolites, tetrahydrocurcumin, hexahydrocurcumin, and octahydrocurcumin. Moreover, electrophoretic mobility shift assay (EMSA) experiments indicated that curcumin blocked the LPS-induced binding of nuclear factor-κB (NFκB), a transcription factor necessary for iNOS induction to its 32P-labeled double-stranded oligonucleotide probe. The inhibition of NFκB activation occurred through the prevention of inhibitor κB (IκB) degradation. Transient transfection experiments also showed that curcumin inhibited NFκB-dependent transcriptional activity. Curcumin blocked the disappearance of inhibitory κBα (IκBα) and p65 from the cytosolic fraction, and inhibited the phosphorylation of IκBα. Furthermore, we showed that curcumin could inhibit the IκB kinase 1 (IKK1) and IκB kinase 2 (IKK2) activities induced by LPS, but tetrahydrocurcumin, hexahydrocurcumin, and octahydrocurcumin were less active. These results suggest that curcumin may exert its anti-inflammatory and anti-carcinogenic properties by suppressing the activation of NFκB through inhibition of IKK activity.