curcumin and neurodegenerative diseases

11
Review Article Curcumin and Neurodegenerative Diseases Adriana Monroy 1 Gordon J. Lithgow 2 Silvestre Alavez 2 * 1 Direcci on de Investigaci on, Hospital General de M exico, M exico D. F., M exico 2 Buck Institute for Research on Aging, Novato, CA, USA Abstract Over the last 10 years curcumin has been reported to be effec- tive against a wide variety of diseases and is characterized as having anticarcinogenic, hepatoprotective, thrombosuppres- sive, cardioprotective, antiarthritic, and anti-infectious proper- ties. Recent studies performed in both vertebrate and invertebrate models have been conducted to determine whether curcumin was also neuroprotective. The efficacy of curcumin in several preclinical trials for neurodegenerative diseases has created considerable excitement mainly because of its lack of toxicity and low cost. This suggests that curcu- min could be a worthy candidate for nutraceutical interven- tion. As aging is a common risk factor for neurodegenerative diseases, it is possible that some compounds that target aging mechanisms could also prevent these kinds of diseases. One potential mechanism to explain several of the general health benefits associated with curcumin is that it may prevent aging-associated changes in cellular proteins that lead to pro- tein insolubility and aggregation. This loss in protein homeo- stasis is associated with several age-related diseases. Recently, curcumin has been found to help maintain protein homeostasis and extend lifespan in the model invertebrate Caenorhabditis elegans. Here, we review the evidence from several animal models that curcumin improves healthspan by preventing or delaying the onset of various neurodegenerative diseases. V C 2013 BioFactors, 39(1):122–132, 2013 Keywords: curcumin; model organisms; mammals; neurodegenerative diseases 1. Introduction Curcumin ((1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6- heptadiene-3,5-dione) is the main component of Curcuma longa, generically known as turmeric, a perennial plant of the family Zingiberaceae that grows naturally in southeast Asia. Turmeric is a spice present in Indian curries and many dishes in south Asia. It has also been used for thousands of years in Indian and Chinese medicine [1]. The main components of tur- meric extracts (curcumin, demethoxycurcumin, and bisdeme- thoxycurcumin) are commonly known as curcuminoids. Multi- ple beneficial effects of curcumin, which could be linked to its ability to act as a strong antioxidant and anti-inflammatory, have been reported during the last 10 years (please see Dr. Shehzad’s paper in this issue). In studies performed on cell cultures and in different animal models, curcumin has been reported to provide a number of beneficial effects. These stud- ies have led to the identification of several pharmacological tar- gets of curcumin and have shed some light on the molecular mechanisms activated by this compound. Interestingly, and maybe because of its lack of toxicity and inexpensive cost, some clinical trials have also been conducted with this compound. Curcumin has been reported to be effective against a wide vari- ety of diseases such as cancer [2,3], cardiovascular disease [4], obesity [5,6], liver disease [7,8], inflammatory disease [9–11], and even aging [12–14] (for a comprehensive review of these topics, please see the appropriate chapters in this issue). In addition to the age-related pathologies noted above, curcumin may have beneficial effects on specific kinds of dis- eases characterized by the formation of aggregated fibrillar proteins deposits. Collectively known as conformational dis- eases [15,16], they are responsible for tremendous social and economic burden. Especially under neurodegenerative condi- tions, the aggregation of aberrant forms of specific proteins such as a-synuclein (Parkinson’s disease, PD) [17], b-amyloid (Alzheimer’s disease, AD) [18], and huntingtin (Huntington’s disease, HD) [19] may contribute to the onset and/or progres- sion of the disease. Initially, the protein aggregates themselves V C 2013 International Union of Biochemistry and Molecular Biology, Inc. Volume 39, Number 1, January/February 2013, Pages 122–132 *Address for correspondence to: Silvestre Alavez, PhD, Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA. Tel.: þ1 415 209 2298; Fax: þ1 415 209 2232; E-mail: [email protected]. Received 27 August 2012; accepted 2 October 2012 DOI: 10.1002/biof.1063 Published online 10 January 2013 in Wiley Online Library (wileyonlinelibrary.com) 122 BioFactors

Upload: silvestre

Post on 14-Dec-2016

216 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Curcumin and neurodegenerative diseases

Review Article

Curcumin and Neurodegenerative Diseases Adriana Monroy1

Gordon J. Lithgow2

Silvestre Alavez2*

1Direcci�on de Investigaci�on, Hospital General de M�exico,M�exico D. F., M�exico2Buck Institute for Research on Aging, Novato, CA, USA

Abstract

Over the last 10 years curcumin has been reported to be effec-

tive against a wide variety of diseases and is characterized as

having anticarcinogenic, hepatoprotective, thrombosuppres-

sive, cardioprotective, antiarthritic, and anti-infectious proper-

ties. Recent studies performed in both vertebrate and

invertebrate models have been conducted to determine

whether curcumin was also neuroprotective. The efficacy of

curcumin in several preclinical trials for neurodegenerative

diseases has created considerable excitement mainly because

of its lack of toxicity and low cost. This suggests that curcu-

min could be a worthy candidate for nutraceutical interven-

tion. As aging is a common risk factor for neurodegenerative

diseases, it is possible that some compounds that target aging

mechanisms could also prevent these kinds of diseases. One

potential mechanism to explain several of the general health

benefits associated with curcumin is that it may prevent

aging-associated changes in cellular proteins that lead to pro-

tein insolubility and aggregation. This loss in protein homeo-

stasis is associated with several age-related diseases.

Recently, curcumin has been found to help maintain protein

homeostasis and extend lifespan in the model invertebrate

Caenorhabditis elegans. Here, we review the evidence from

several animal models that curcumin improves healthspan by

preventing or delaying the onset of various neurodegenerative

diseases. VC 2013 BioFactors, 39(1):122–132, 2013

Keywords: curcumin; model organisms; mammals; neurodegenerative

diseases

1. Introduction

Curcumin ((1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) is the main component of Curcumalonga, generically known as turmeric, a perennial plant of thefamily Zingiberaceae that grows naturally in southeast Asia.Turmeric is a spice present in Indian curries and many dishesin south Asia. It has also been used for thousands of years inIndian and Chinese medicine [1]. The main components of tur-meric extracts (curcumin, demethoxycurcumin, and bisdeme-thoxycurcumin) are commonly known as curcuminoids. Multi-ple beneficial effects of curcumin, which could be linked to itsability to act as a strong antioxidant and anti-inflammatory,

have been reported during the last 10 years (please see Dr.Shehzad’s paper in this issue). In studies performed on cellcultures and in different animal models, curcumin has beenreported to provide a number of beneficial effects. These stud-ies have led to the identification of several pharmacological tar-gets of curcumin and have shed some light on the molecularmechanisms activated by this compound. Interestingly, andmaybe because of its lack of toxicity and inexpensive cost, someclinical trials have also been conducted with this compound.Curcumin has been reported to be effective against a wide vari-ety of diseases such as cancer [2,3], cardiovascular disease [4],obesity [5,6], liver disease [7,8], inflammatory disease [9–11],and even aging [12–14] (for a comprehensive review of thesetopics, please see the appropriate chapters in this issue).

In addition to the age-related pathologies noted above,curcumin may have beneficial effects on specific kinds of dis-eases characterized by the formation of aggregated fibrillarproteins deposits. Collectively known as conformational dis-eases [15,16], they are responsible for tremendous social andeconomic burden. Especially under neurodegenerative condi-tions, the aggregation of aberrant forms of specific proteinssuch as a-synuclein (Parkinson’s disease, PD) [17], b-amyloid(Alzheimer’s disease, AD) [18], and huntingtin (Huntington’sdisease, HD) [19] may contribute to the onset and/or progres-sion of the disease. Initially, the protein aggregates themselves

VC 2013 International Union of Biochemistry and Molecular Biology, Inc.Volume 39, Number 1, January/February 2013, Pages 122–132*Address for correspondence to: Silvestre Alavez, PhD, Buck Institute forResearch on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.Tel.: þ1 415 209 2298; Fax: þ1 415 209 2232;E-mail: [email protected] 27 August 2012; accepted 2 October 2012DOI: 10.1002/biof.1063Published online 10 January 2013 in Wiley Online Library(wileyonlinelibrary.com)

122 BioFactors

Page 2: Curcumin and neurodegenerative diseases

were considered to be the toxic insult leading to cell death.However, more recent studies suggest that soluble aggregateprecursors such as soluble oligomers or fibrils may initiate pa-thology by influencing neuronal function [15]. Although adetailed characterization of how these insoluble intracellularand extracellular deposits develop is still unclear, multiple fac-tors such as pH, metal ions, protein concentration, and oxida-tive stress have been reported to play a role in theirformation.

It has been estimated that AD and PD are the twoneurodegenerative diseases with the greatest incidence in theUSA and that AD accounts for 60–80% of all dementiadiagnosed [20]. The costs associated with treating thesediseases and paying caregivers, as well as the loss of produc-tivity of those afflicted, in 2010 has been estimated to be �800billion Euros [21]. These statistics strongly suggest that devel-oping therapies aimed to reduce or delay neurodegenerativedisease should be a priority for the biomedical community.

PD is named after James Parkinson who described ‘‘shak-ing palsy’’ in 1817. This disease is characterized by severalsymptoms such as bradykinesia, tremors, rigidity, dementia,and depression. At the molecular level, this motor dysfunctionhas been associated with pathological spherical inclusions inneurons of the substantia nigra, known as Lewy bodies, andwith a loss of nigrostriatal dopamine (DA) neurons. Despitegreat effort, the etiology of this disease is still undetermined.However, it does seem clear that a cumulative loss of dopami-nergic neurons during aging could contribute to the onset/pro-gression of the disease. This pathology can be accelerated byexposure to environmental toxins, excitotoxicity, oxidativestress, or mutations in the a-synuclein gene, which encodes aprotein found in Lewy bodies in idiopathic PD lesions. Becauseof the loss of dopaminergic neurons in the substantia nigra,striatal cholinergic neurons are disinhibited, leading to animbalance of dopaminergic/cholinergic neurotransmission [22].As DA does not cross the blood–brain barrier, pharmacologicalapproaches to treat PD have focused on developing drugs ableto increase DA or reduce acetylcholine (ACh) activity in thebrain. The primary approaches include the use of levodopa, aDA precursor and indirect agonist of G protein-coupled D2receptors, which increase DA production; direct stimulation ofD2 receptors with selective agonists such as bromocriptine,ropinirole, pramipexole, or rotigotine; or preventing DA enzy-matic catabolism by enzymes such as monoamine oxidase typeB (e.g., selegiline and rasagiline) or catechol-O-methyltransfer-ase (e.g., entacapone and tocalpone). These compounds are of-ten administered with antimuscarinics, such as benztropine ortrihexyphenidyl, to decrease striatal cholinergic excitability[23].

AD is a neurodegenerative illness characterized by earlyonset of short-term memory loss and cognitive decline thateventually leads to dementia. This disease is associated with aparticular brain pathology that includes neurofibrillar tanglesand senile plaques (b-amyloid, Ab). Aggregated Ab in senileplaques have a b sheet secondary structure and are arranged

in fibrils [24]. A significant amount of neuronal loss has beenreported during AD progression (e.g., basal forebrain, hippo-campus, and associative cerebral cortex). At molecular level,this neuronal loss seems to be associated with a reduction ofcholine acetyltransferase activity and, as a consequence, witha marked diminution in ACh levels. No drugs are currentlyavailable to prevent this neuronal degeneration. To date, thepotential antiamyloid therapeutic approaches to treat AD focuson the amyloid cascade theory, such as the Ab vaccine ortreatment with metal-complexing agents [25,26]. However,several drugs that prevent ACh degradation have also beenused to improve cognition during AD (e.g., tacrine ordonepezil).

HD is a neurodegenerative disorder caused by the autoso-mal dominant mutation of the huntingtin gene. Altered proteinaggregates affect muscle coordination and lead to abnormalinvoluntary movements, known as chorea, as well as cognitiveand psychiatric problems. Tetrabenazine, an inhibitor of thevesicular monoamine transporter 2 (VMT2) that promotes do-pamine degradation, is used to treat Huntington’s chorea, butnot to treat HD itself [27].

Unfortunately, current treatments in PD, AD, and HD,beyond symptomatic improvement, do not have neuroprotec-tive properties or the potential to modify the course of the dis-ease, and even symptomatic relief is temporary. Additionally,all these compounds are highly toxic and can cause severeside effects (nauseas, stomach cramps, dizziness, drowsiness,insomnia, headache, diarrhea, dry mouth, mydriasis and evendelirium, depression, or hallucinations).

2. An Important Role for Curcumin

With all this in mind, it is easy to understand the excitementgenerated by a compound like curcumin. If curcumin could beshown to have strong efficacy, it has the potential to become acandidate for nutraceutical intervention in neurodegenerativedisease. It is interesting to note that, because of its strong af-finity for fibrillar amyloid proteins, curcumin is already usedto stain in vitro tissue sections from affected individuals [28].The search for curcumin derivatives with higher specificitiesfor Ab fibrils and adequate lipophilic properties for crossingthe blood–brain barrier is a subject of current research[29,30]. Further fueling these efforts is research showing thatcurcumin is able to prevent aggregation of Ab in vitro and incell cultures [31,32], suggesting that curcumin could alter theeffects of protein aggregation in animal models and potentiallyin humans.

However, as alluded earlier, one of the principal limita-tions for the use of curcumin in nutraceutical interventions isits limited bioavailability, which is mainly because of its poorabsorption and fast metabolism. Although curcumin is verystable in acidic media, at physiological pH it is easily degradedto ferulic acid and feruloylmethane [33]. Whether these metab-olites could have similar properties to those reported for

Monroy et al. 123

Page 3: Curcumin and neurodegenerative diseases

curcumin is still an active field of research. In parallel, effortsto increase its bioavailability in mammals, particularly inhumans [34–37], by conjugating it to a stable carrier or bycoadministering it with inhibitors of curcumin metabolismhave rendered some interesting results. Research in this areais comprehensively covered in Dr. Helson’s review in this spe-cial issue.

Despite what are apparent pharmacokinetic limitations,curcumin has been reported to have multiple pharmacologicalactivities and to be effective against a wide variety of diseasesbecause of its anticarcinogenic [2,3,7,37–40], hepatoprotective[8,41–44], thrombosuppressive [45,46], cardioprotective [47–49], antiarthritic [9–11], and anti-infectious properties [50–54].

Everything considered, the demographic shift toward anolder population makes compounds with this broad spectrumof potential clinical applications particularly interesting. Theremainder of this review will summarize the effects of curcu-min in diverse experimental models of neurodegenerative dis-eases and speculate on the directions the field is headed inthe immediate future. We particularly emphasize studies ofcurcumin in invertebrate models, mice and clinical trials inhumans.

3. Effect of Curcumin in Cell Cultures

In addition to the reported benefits of curcumin in traditionalChinese and Indian medicine, the beneficial effects of curcu-min have been demonstrated in a wide variety of cells, includ-ing neurons [55], astrocytes [56], and microglia [57]. Effectshave also been tested in primary cell cultures from differentregions of the central nervous system, including cerebral cor-tex [58], mesencephalon [59], hippocampus [55], and spinalcord [60]. Curcumin is known to possess neuroprotective prop-erties [61], and its anti-inflammatory [62], antioxidant [63],and insulin-sensitizing effects [64] have been described usingneuronal/glial primary and immortalized cells though its inter-action with different molecular targets including metals, proin-flammatory cytokines, protein kinases, and other enzymes[65].

Ab aggregation is a feature of AD and curcumin has beenshown to be able to inhibit the formation of the Ab fibrils invitro [66]. In fact, in a classic experiment where 214 antioxi-dant compounds were tested, curcumin proved the strongestinhibitor effect on the formation of Ab fibrils [67]. Further-more, curcumin demonstrates a dose-dependent effect on theinhibition of Ab1–40/1–42 fibrils and even destabilizes preformedfibrils in vitro [66]. Several studies have also demonstrated theability of some curcuminoid compounds, including turmericextract, to suppress Ab aggregation and oligomerization [68–70]. Yang et al. documented the ability of curcumin to inhibitAb aggregation and protect against Ab-induced cell death [31].In addition, Zhang et al. showed that curcumin decreased bothAb levels and amyloid precursor protein (APP) maturation in

mouse primary cortical neurons [71]. Curcumin-mediated cellsurvival in Ab as well as APP-challenged cell systems is due toattenuation of apoptosis and oxidative injury. Curcumin treat-ment of human neuroblastoma SK-M-NC cells prevents celldeath elicited by the Ab peptide through the inhibition of NFjBactivation [72]. Similarly, PC12 rat pheochromocytoma cellsare protected from Ab insult through an antioxidant pathway[73]. Interestingly, curcumin is also able to prevent the fibrilla-tion pattern of a-synuclein, the main protein involved in PD,in vitro [74].

Neuroinflammation plays a key role in the onset and pro-gression of neurodegenerative diseases. In line with this idea,curcumin reduces the expression of IL-1a, IL-6, and TNF-a inLPS-stimulated BV2 microglia in a dose-dependent manner[75]. It is known that amyloid aggregates can be cleared viaphagocytosis by brain macrophages and that patients with ADshow signs of defective phagocytosis that results in an ineffec-tive clearance of Ab plaques. Interestingly, curcumin can stim-ulate microglial phagocytosis and clearance of Ab in vitro aswell as increase the induction of heat-shock proteins inresponse to the addition of soluble Ab aggregates to neuronalcultures [76].

A link between iron metabolism and AD pathogenesis issuggested by the presence of an iron-responsive element in the50 UTR of the APP mRNA [77]. Additionally, high amounts ofiron and copper in amyloid plaques could be responsible forstimulating free radical generation and thus increasing proteinand DNA oxidation, lipid peroxidation, advanced glycation endproducts, carbonyls, malondialdehyde, peroxynitrite, and hemeoxygenase-1 (OH-1) while decreasing levels of cytochrome c oxi-dase [78]. In this context, the antioxidant properties of curcu-min might also be linked to its capacity to complex with redox-active metals because curcumin can bind iron or copper ions inin vitro experiments [79] and because copper–curcumin com-plexes show radical scavenger and superoxide dismutase-likeproperties [80].

PD has been modeled in vitro through the specificneurotoxic effect of the 6-hydroxydopamine (6-OHDA) and1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [81] ondopaminergic neurons. Neurotoxicity triggered by 6-OHDA isattenuated by curcumin treatment in both SH-SY5Y andMES23.5 cells through the inhibition of reactive oxygenspecies (ROS), mitochondrial protection, and antiapoptoticmechanisms [82,83]. Curcumin also confers protection againstMPTP- (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) andMPPþ (1-methyl-4-phenylpyridinium)-induced apoptosis inPC12 cells through the Bcl-2-mitochondria-ROS-iNOS pathway[84] and by the inhibition of the JNK pathway, which contrib-utes to preventing dopaminergic neuronal death in SH-SY5Ycells [85].

Taken together, these results suggest that curcumin couldalso have beneficial effects in whole organisms. In the nextsection, we will analyze the current state of research involvingcurcumin and invertebrate models of aging and neurodegener-ative disease.

BioFactors

124 Neuroprotective Effects of Curcumin

Page 4: Curcumin and neurodegenerative diseases

4. Effect of Curcumin in InvertebratesModels of Aging and Neurodegeneration

Caenorhabditis elegans has been used to identify moleculeswith a broad range of activities such as antihelminthics, anti-fungals, and compounds with neuronal activity [86–91]. Addi-tionally, a number of pharmacological interventions on theaging process have been demonstrated in this system [92–105]. Many of the C. elegans small-molecule studies to datehave focused on the effects of natural products such asextracts from Ginkgo biloba [106], blueberry phenols [107], orresveratrol [108]. Interestingly, curcumin is also able toincrease lifespan through a mechanism that involves theregulation of protein homeostasis [104]. However, curcuminseems to have no effect on mouse lifespan [109], although it ispossible that this compound could affect other aspects of agingincluding neurodegenerative diseases or that it could extendlifespan when administered differently.

In addition to the multiple benefits and resources for ex-ploitation of the worm system as a platform for drug discovery,worms have also been genetically engineered to expresshuman disease-associated proteins. For example, a robustmodel of protein aggregation in which human Ab peptide3–42 isexpressed under the control of the unc-54 myosin promoter inmuscle tissue has been developed in C. elegans [110]. Modelsexpressing tau protein in neurons [111,112], a-synuclein in dif-ferent tissues [113–116], and mouse prion protein [117] havealso been generated in this model system. There are alsotransgenic worms expressing chains of polyQ of differentlengths, characteristic of the huntingtin protein, tagged to YFPdriven by the unc-54 promoter. As observed in humans, theydemonstrate an aggregation rate that is dependent on thepolyQ repeat length, which culminates in a paralysis pheno-type [118,119]. Several compounds that increase lifespan havebeen reported to prevent protein aggregation in these models[120]. Recently, we exploited some of these worm models ofneurodegenerative diseases to test several compounds that weidentified as having prolongevity properties [104]. We foundthat these compounds significantly decreased the paralysisphenotype associated with Ab peptide3–42, PolyQ, myosin, andperlecans aggregation through a mechanism that requirescomponents of the protein homeostasis network. In particular,we found that 100 lM curcumin supplementation increasesworm lifespan by a mechanism that depends on both the heatshock factor 1 (HSF-1), a transcription factor that has longbeen associated with the control of stress resistance, as wellas a dietary restriction (DR)-like mechanism [121]. Theseresults were later confirmed by another laboratory that foundthat curcumin’s antioxidant properties are also required forthe lifespan increase induced by this compound [13].

Curcumin also increases lifespan, enhances stressresistance, and improves spontaneous locomotion in two dif-ferent strains of Drosophila melanogaster [12]. Similar to thecase in worms, Drosophila has been genetically engineered toproduce models of neurodegenerative disease including AD,

PD, tauopathies, several polyglutamine disorders, amyotrophiclateral sclerosis, and Prion disease [122]. Unfortunately, just afew interventions in neurodegenerative diseases are found inthe literature. Recently, Caesar et al. showed that curcumin(1–100 lg/g yeast paste) is able to increase lifespan andimprove locomotion in five different genetic models of AD inDrosophila [123]. After curcumin treatment, they found nochanges in number or size of Ab deposits but instead observeda tendency to favor amyloid fibril formation over the solubleoligomeric Ab species.

These studies demonstrate that a process long associatedwith age-related neurodegenerative disease is also a generalfeature of aging, suggesting that the loss of protein homeosta-sis could be a common mechanism of aging and disease. It fol-lows that pharmacologically targeting the age-related declineof protein homeostasis could reduce and/or postpone neurode-generative disease and extend lifespan.

There are, of course, many limitations to using these sim-ple animal models and we should not underestimate the com-plexity of human disease by comparing to the events modeledin worms and flies. Indeed, most of the enzymatic machinerythat processes neurotoxic proteins is not present in theseinvertebrates, and in most cases they lack critical modulatorsof disease progress such as complex inflammatory responses.

Nevertheless, these results suggest that edible compoundslike curcumin could be used to suppress age-related diseasepathologies and underline some important concepts for a novelapproach to the discovery of new drugs with the potential toimprove the conditions of neurodegenerative diseases. It isalso important to consider that compounds that affect the dy-namics and patterns of protein aggregation could also gener-ate some soluble oligomeric species potentially toxic to thecell. Additionally, at the concentration required to affect pro-tein aggregation, some compounds could have off-targeteffects that act in parallel to produce deleterious effects on cellphysiology. Therefore, caution ought to be taken when consid-ering compounds identified through this experimentalapproach for use as potential therapeutic drugs.

5. Effect of Curcumin in Mammals

A small number of compounds have been tested for their abil-ity to increase lifespan and improve healthspan in mammals.Among others, nordihydroguaiaretic acid and aspirin (2-ace-toxybenzoic acid) increase the lifespan of male, but notfemale mice [124], probably because of sex-specific pharma-cokinetics of these drugs. Interestingly, the immunosuppres-sant rapamycin that inhibits mTOR signaling is able toincrease lifespan in both male and female mice when admin-istered late in life [125,126]. Therefore, it is reasonable toassume that compounds that positively affect lifespan couldalso produce beneficial effects on degenerative diseases andhence, the effect of these compounds on healthspan is anactive field of research.

Monroy et al. 125

Page 5: Curcumin and neurodegenerative diseases

Despite there being no evidence that curcumin affects life-span in either male or female mice when administered begin-ning at 4 months of age [109], several interesting studies inrodents have examined the ability of curcumin to provide neu-roprotection from neurodegenerative disorders, especially ADand PD. For a review of the effect of curcumin in aging, pleasesee Dr. Lai’s chapter in this issue. In a pioneering study,Frautschy et al. [127] showed that a curcumin nutraceuticalintervention attenuated oxidative injury, microgliosis, synapto-physin loss, spatial memory deficits, postsynaptic loss, and Abdeposits produced by intracerebroventricular infusion of Abamyloid in rats.

The Ab-overexpressing Tg2576 APPSw transgenic mouse,a popular mouse model for AD, has also been used as a pre-clinical tool to evaluate the neuroprotective potential of curcu-minoids on this disease. Lim et al. [128] evaluated the effecton Tg2576 mice fed for 6 months with high doses (5,000 ppm)or low doses (160 ppm) of curcumin and found that both dosessignificantly decrease two biochemical conditions which nor-mally are found elevated in the brains of these mice, proteinoxidation and the levels of the proinflammatory cytokine inter-leukin-1b. In their experiment, low doses of curcumin attenu-ated Ab overexpression, decreased oxidative damage, and alsoreduced levels of glial fibrillary acidic protein, which isinvolved in brain injury and inflammation. In a subsequentstudy in Tg2576 mice, dietary administration of curcuminreduced amyloid plaque formation, attenuated both ROS andreactive nitrogen species (NOS) formation, as well asdecreased cell death [129]. Interestingly, in a study where cur-cumin was intravenously administered in Ab-overexpressingmice (PS1dE9), beneficial effects including plaque disruptionand attenuation of distorted neuritis were also observed [28].Besides its antiamyloid, antioxidant, anti-inflammatory, andcholesterol-lowing properties, curcumin can also attenuatememory deficits in AlCl3 and D-galactose-challenged Kunmingmice [130].

Taken together, these studies illustrate the potential ofcurcumin to reverse neurodegeneration and improve thecognitive impairments associated with AD. There are also datasuggesting that curcumin could have a protective effect againstneurodegeneration in PD. As found for Ab, curcumin can alsoinhibit the aggregation of a-synuclein [131], and several stud-ies performed in rodents have examined the neuroprotectivepotential of curcuminoids against both MPTP- and 6-OHDA-induced dopaminergic degeneration. In these studies, orallyand intravenously administered curcumin modulates dopami-nergic damage in 6-OHDA-treated rodents by suppressing apo-ptosis and inducing microglial activation with a consequentimprovement in locomotion [132,133]. Dietary, intravenous,and intraperitoneal curcumin administration reversed the do-paminergic neurotoxicity in MPTP-treated rodents. In theseexperiments, curcumin was able to decrease the oxidativestress, inhibit the activity of monoamine oxidase B, suppressapoptosis, inhibit protein nitration, increase levels of glutathi-one, and decrease the activity of mitochondrial complex I

induced by MPTP treatment [134–137]. Unfortunately, thereare limited data on the potential beneficial role of curcumin inother neurodegenerative diseases. This could be the result ofthe low incidence of these diseases. Recently, it has beenshown that dietary curcumin in a dose of 555 ppm causes adecrease in huntingtin protein aggregation, improved rearingdeficits, but impairing climbing behavior in the CAG140 mice,an animal model of HD [138]. These in vivo preclinical studiesexhibit the potential of curcumin as a neuroprotector and sug-gest a role for this compound in the prevention and reversal ofdegenerative diseases such as AD or PD (Table 1).

6. Curcumin in Clinical Trials

As described above, therapeutic options for the treatment ofneurodegenerative diseases offer limited benefits and multipleside effects, indicating the need to develop safe and effectivepharmacological agents for the prevention and treatment ofthese kinds of diseases. Because of its antioxidant and anti-inflammatory effects, as well as its ability to inhibit proteinaggregation, curcumin represents one of the most promisingcompounds with therapeutic potential. Epidemiologic evidenceof curcumin action is illustrated in a recent large population-based study of 1,010 elderly nondemented Asians. Subjects inthis study that consumed curry occasionally often or very oftenscored significantly better on the Mini-Mental State Examina-tion, an established measure of cognitive function, than didthose who never or rarely consumed curry [139]. The thera-peutic use of curcumin has been tested in two independentclinical trials. The first one was a 6-month randomized, pla-cebo-controlled, double-blind, clinical pilot study of curcuminconducted in patients with AD in Hong Kong, China. In thisstudy, 34 subjects started the trial and 27 completed; 8 sub-jects on 0 g, 9 on 1 g, and 11 on 4 g curcumin per day. No dif-ference was observed between the 1 and 4 g groups. On thisstudy, curcumin serum levels reached a maximum of 250 nMat 1.5 h when given with food and 270 nM at 4 h when givenwith water. The inability to detect any relative protective effectof curcumin could be due to the lack of cognitive decline in theplacebo group. However, when compared with the placebocontrol group, curcumin showed increased plasma levels ofvitamin E and increased serum Ab40, suggesting that curcumincould disaggregate Ab deposits in the brain and release Ab forcirculation and disposal [140]. The second clinical trial was aphase II double-blind study on mild to moderate AD, per-formed in CA, USA, with patients that receive 2–10 mg of cur-cumin for 6 months. Unfortunately, no significant improvementin cognitive function or changes in the levels of Ab, total tau,and phosphorylated tau in plasma and CSF were found [141](Table 1). To date, two other studies are still active. One is aphase II study in India using 2 g/day of curcumin. The second isan early intervention conducted in the USA with a combinationof 5.4 g of curcumin and bioperine, another natural productderived from black pepper that has been claimed to increase

BioFactors

126 Neuroprotective Effects of Curcumin

Page 6: Curcumin and neurodegenerative diseases

the bioavailability of several nutritional compounds. These stud-ies are directed to evaluate the efficacy, safety, and tolerabilityof curcumin in moderate AD.

Despite the somewhat disappointing results of the clinicaltrials available to date, it is premature to conclude a total lack ofeffect of curcumin on AD or PD. Additional studies with largernumbers of patients and longer period of treatment could berequired to improve the clinical conditions, delay the onset, orameliorate the progression of these diseases.

7. Conclusions and Future Directions

Although identifying compounds that improve the healthspanof mammals is undoubtedly more relevant for human drug de-velopment, the prohibitive cost of mouse studies makes itextremely unlikely that large-scale chemical screens will becarried out in mice. Basic research in more cost-effectivemodel systems is therefore a critical starting point for identify-ing such compounds and elucidating their mechanism(s) ofaction. Cell culture and invertebrate model organisms provideopportunities to assay promising compounds, like curcumin, inan efficient manner. Moreover, once candidate compounds areidentified, model systems allow for rapid elucidation of thegenetic pathways being targeted by these compounds. Further-

more, some pathological features of certain diseases are nowbeing seen as a more general feature of aging. Perhaps, theclearest example of this is the failure of protein homeostasisassociated with age-related neurological disease, which leadsto the formation of intracellular or extracellular protein aggre-gates. This is consistent with a mechanistic relationshipbetween aging and disease.

The disappointing outcomes of dozens of phase III clinicaltrials in AD, PD, and others suggest that preclinical studies inanimal models are less relevant than we would hope. As agingis a major risk factor for many human diseases, compoundsthat slow aging are highly sought- after because of their poten-tial for treating age-related diseases. Here, we argue that therecent growth of a new subfield, the chemical biology of aging,will lead to the identification of candidate compounds andmechanistic insights that will ultimately propel forward treat-ments of age-related diseases. Curcumin is a great example ofthis general idea because of the multiple beneficial effectsreported for curcumin in vitro. Curcumin has been reported toincrease lifespan in C. elegans and Drosophila but does notseem to increase lifespan in mice. Nevertheless, there isenough evidence to suggest that curcumin could be of help inthe treatment of several neurodegenerative diseases and otherage-associated diseases to significantly improve healthspan.This could be due to its well-known antioxidant and anti-

Preclinical and clinical studies described in this paper

Animal model Dose Disease Effects Reference

Sprague-Dawley rats Diet, 500 and 2,000 ppm,

2 months

AD (Ab ICV infusion) ;Spatial memory deficits, oxidative

damage, microgliosis, synaptophysin

127

Tg2576 APPSw mice Diet, 160 and 5,000 ppm,

6 months

AD (Ab overexpression) ;IL-1b, oxidative damage, GFAP, Ab 128

Tg2576 APPSw mice Diet, 500 ppm, 4 months AD (Ab overexpression) ;Cell death, iNOS, IL-1b 129

PS1dE9 mice IV, 7.5 mg/kg/day, 7 days AD (Ab overexpression) :Restoration of distorted neuritis,

plaque disruption

28

Kunming mice PO, 200 mg/kg, 45 days AD(AlCl3, D-galactose) ;Spatial memory deficits 130

Sprague-Dawley rats PO, 50 mg/kg, 4 days PD (6-OHDA) THþ cells protection 132

ICR mice IP, 50 mg/kg, 3 times PD (MPTP) ;Oxidative damage, :dopaminergic

neurons

134

Swiss albino mice IP, 80 mg/kg, 7 days PD (MPTP) ;MAO-B 136

CAG140 Diet, 555 ppm HD ;Protein aggregation, :rearing, ;climbing 138

Humans Curry consumption AD Improve in cognitive function 139

Humans 1–4 g AD No changes in cognitive function 140

Humans 2–10 mg AD No changes in cognitive function or

Ab serum levels

141

TABLE 1

Monroy et al. 127

Page 7: Curcumin and neurodegenerative diseases

inflammatory properties, but could be also the result of a mod-ulation of protein aggregation through the regulation of pro-tein homeostasis or a dietary restriction-like mechanism asrecent studies in worms suggest (Fig. 1). As most of the drugtherapies for neurodegenerative diseases (like AD and PD) cur-rently available have shown little efficacy and produce multipleside effects, a nutraceutical intervention with an innocuous andcheap compound like curcumin could represent a major avenuefor the treatment of these diseases. One of the main limitationsfor a nutraceutical intervention with curcumin on neurodege-nerative diseases is its limited bioavailability. This could beaddressed by chemical modifications of curcumin, through itsconjugation with lipophilic compounds or by coadministration ofcurcumin with compounds that facilitate its absorption. Despitethat, clinical trials available to date do not provide conclusiveevidence of the efficacy of curcumin for preventing or treatingneurodegenerative diseases. There are, however, some encour-aging results suggesting that curcumin could be of therapeuticrelevance in these kinds of diseases. Of course, more studies areneeded to explore the effects of this compound in long-termnutraceutical interventions and the fact that curcumin seems tobe innocuous in humans could prompt additional studies on the

effect of curcumin in the onset and progression of several neuro-degenerative diseases.

AcknowledgementsThe authors thank Aric N. Rogers for helpful discussion. G.J.L.is supported by the NIH AG21069, AG22868, AG029631-01A1,ES016655, the Larry L. Hillblom Foundation, and UL1RR024917. S.A. was supported by the U19AGO231222 fromthe Longevity Consortium.

References

[1] Prasad, S. and Aggarwal, B. B. In Benzie, I. F. F., Wachtel-Galor, S., Eds.

(2011) Turmeric, the golden spice: from traditional medicine to modern

medicine, CRC Press, Boca Raton.

[2] Basnet, P. and Skalko-Basnet, N. (2011) Curcumin: an anti-inflammatory

molecule from a curry spice on the path to cancer treatment. Molecules

16, 4567–4598.

[3] Darvesh, A. S., Aggarwal, B. B., and Bishayee, A. (2012) Curcumin and

liver cancer: a review. Curr. Pharm. Biotechnol. 13, 218–228.

[4] Jagtap, S., Meganathan, K., Wagh, V., Winkler, J., Hescheler, J., et al.

(2009) Chemoprotective mechanism of the natural compounds,

Model depicting the proposed mechanism of curcumin to provide neuroprotection. By it chemical structure, curcumin may act as

a natural free radical (ROS) scavenger. Acting through the neurotrophic factor jB (NF-jB) curcumin can decrease the release of

different interleukins. Curcumin could act as a stress response mimetic that induces some components of the protein homeosta-

sis network or as it is known to bind amyloid, directly acts in the misfolded cascade. This induction requires the transcription fac-

tors HSF-1 and Nrf2 (SKN-1 in C. elegans). Additionally, curcumin could act as a DR mimetic to activate these transcription factors

through the AMPK pathway (Black arrows ¼ induction, red symbol ¼ inhibition).

FIG 1

BioFactors

128 Neuroprotective Effects of Curcumin

Page 8: Curcumin and neurodegenerative diseases

epigallocatechin-3-O-gallate, quercetin and curcumin against cancer and

cardiovascular diseases. Curr. Med. Chem. 16, 1451–1462.

[5] Aggarwal, B. B. (2010) Targeting inflammation-induced obesity and meta-

bolic diseases by curcumin and other nutraceuticals. Annu. Rev. Nutr. 30,

173–199.

[6] Alappat, L. and Awad, A. B. (2010) Curcumin and obesity: evidence and

mechanisms. Nutr. Rev. 68, 729–738.

[7] Sharma, R. A., Ireson, C. R., Verschoyle, R. D., Hill, K. A., Williams, M. L.,

et al. (2001) Effects of dietary curcumin on glutathione S-transferase and

malondialdehyde-DNA adducts in rat liver and colon mucosa: relationship

with drug levels. Clin. Cancer Res. 7, 1452–1458.

[8] El-Agamy, D. S. (2010) Comparative effects of curcumin and resveratrol on

aflatoxin B(1)-induced liver injury in rats. Arch. Toxicol. 84, 389–396.

[9] Funk, J. L., Frye, J. B., Oyarzo, J. N., Kuscuoglu, N., Wilson, J., et al.

(2006) Efficacy and mechanism of action of turmeric supplements in the

treatment of experimental arthritis. Arthritis Rheum. 54, 3452–3464.

[10] Funk, J. L., Oyarzo, J. N., Frye, J. B., Chen, G., Lantz, R. C., et al. (2006)

Turmeric extracts containing curcuminoids prevent experimental rheuma-

toid arthritis. J. Nat. Prod. 69, 351–355.

[11] Chandran, B. and Goel, A. (2012) A randomized, pilot study to assess the

efficacy and safety of curcumin in patients with active rheumatoid arthritis.

Phytother. Res. 26, 1719–1725.

[12] Lee, K. S., Lee, B. S., Semnani, S., Avanesian, A., Um, C. Y., et al. (2010)

Curcumin extends life span, improves health span, and modulates the

expression of age-associated aging genes in Drosophila melanogaster.

Rejuvenation Res. 13, 561–570.

[13] Liao, V. H., Yu, C. W., Chu, Y. J., Li, W. H., Hsieh, Y. C., et al. (2011)

Curcumin-mediated lifespan extension in Caenorhabditis elegans. Mech.

Ageing Dev. 132, 480–487.

[14] Pallauf, K. and Rimbach, G. (2012) Autophagy, polyphenols and healthy

ageing. Ageing Res. Rev. 12, 237–252.

[15] Kopito, R. R. and Ron, D. (2000) Conformational disease. Nat. Cell Biol. 2,

E207–E209.

[16] Kikis, E. A., Gidalevitz, T., and Morimoto, R. I. (2010) Protein homeostasis

in models of aging and age-related conformational disease. Adv. Exp.

Med. Biol. 694, 138–159.

[17] Selkoe, D. J. (2003) Folding proteins in fatal ways. Nature 426, 900–904.

[18] Haass, C. and Selkoe, D. J. (2007) Soluble protein oligomers in neurode-

generation: lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev.

Mol. Cell Biol. 8, 101–112.

[19] Bates, G. (2003) Huntingtin aggregation and toxicity in Huntington’s dis-

ease. Lancet 361, 1642–1644.

[20] Nussbaum, R. L. and Ellis, C. E. (2003) Alzheimer’s disease and Parkin-

son’s disease. N. Engl. J. Med. 348, 1356–1364.

[21] Gustavsson, A., Svensson, M., Jacobi, F., Allgulander, C., Alonso, J., et al.

(2011) Cost of disorders of the brain in Europe 2010. Eur. Neuropsycho-

pharmacol. 21, 718–779.

[22] Meissner, W. G., Frasier, M., Gasser, T., Goetz, C. G., Lozano, A., et al.

(2011) Priorities in Parkinson’s disease research. Nat. Rev. Drug Discov.

10, 377–393.

[23] Schapira, A. H., Bezard, E., Brotchie, J., Calon, F., Collingridge, G. L., et al.

(2006) Novel pharmacological targets for the treatment of Parkinson’s dis-

ease. Nat. Rev. Drug Discov. 5, 845–854.

[24] Ittner, L. M. and Gotz, J. (2011) Amyloid-beta and tau--a toxic pas de deux

in Alzheimer’s disease. Nat. Rev. Neurosci. 12, 65–72.

[25] Finefrock, A. E., Bush, A. I., and Doraiswamy, P. M. (2003) Current status

of metals as therapeutic targets in Alzheimer’s disease. J. Am. Geriatr.

Soc. 51, 1143–1148.

[26] Barnham, K. J. and Bush, A. I. (2008) Metals in Alzheimer’s and

Parkinson’s diseases. Curr. Opin. Chem. Biol. 12, 222–228.

[27] Ha, A. D. and Fung, V. S. (2012) Huntington’s disease. Curr. Opin. Neurol.

25, 491–498.

[28] Garcia-Alloza, M., Borrelli, L. A., Rozkalne, A., Hyman, B. T., and Bacskai,

B. J. (2007) Curcumin labels amyloid pathology in vivo, disrupts existing

plaques, and partially restores distorted neurites in an Alzheimer mouse

model. J. Neurochem. 102, 1095–1104.

[29] Ran, C., Xu, X., Raymond, S. B., Ferrara, B. J., Neal, K., et al. (2009)

Design, synthesis, and testing of difluoroboron-derivatized curcumins as

near-infrared probes for in vivo detection of amyloid-beta deposits. J. Am.

Chem. Soc. 131, 15257–15261.

[30] Lee, I., Yang, J., Lee, J. H., and Choe, Y. S. (2011) Synthesis and evalua-

tion of 1-(4-[(1)(8)F]fluoroethyl)-7-(40-methyl)curcumin with improved brain

permeability for beta-amyloid plaque imaging. Bioorg. Med. Chem. Lett.

21, 5765–5769.

[31] Yang, F., Lim, G. P., Begum, A. N., Ubeda, O. J., Simmons, M. R., et al.

(2005) Curcumin inhibits formation of amyloid beta oligomers and fibrils,

binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 280,

5892–5901.

[32] Hamaguchi, T., Ono, K., Murase, A., and Yamada, M. (2009) Phenolic com-

pounds prevent Alzheimer’s pathology through different effects on the

amyloid-beta aggregation pathway. Am. J. Pathol. 175, 2557–2565.

[33] Wang, Y. J., Pan, M. H., Cheng, A. L., Lin, L. I., Ho, Y. S., et al. (1997) Sta-

bility of curcumin in buffer solutions and characterization of its degrada-

tion products. J. Pharm. Biomed. Anal. 15, 1867–1876.

[34] Anand, P., Kunnumakkara, A. B., Newman, R. A., and Aggarwal, B. B.

(2007) Bioavailability of curcumin: problems and promises. Mol. Pharm. 4,

807–818.

[35] Gota, V. S., Maru, G. B., Soni, T. G., Gandhi, T. R., Kochar, N., et al. (2010)

Safety and pharmacokinetics of a solid lipid curcumin particle formulation

in osteosarcoma patients and healthy volunteers. J. Agric. Food Chem. 58,

2095–2099.

[36] Kumar, A., Ahuja, A., Ali, J., and Baboota, S. (2010) Conundrum and thera-

peutic potential of curcumin in drug delivery. Crit. Rev. Ther. Drug Carrier

Syst. 27, 279–312.

[37] Yallapu, M. M., Jaggi, M., and Chauhan, S. C. (2012) Curcumin nanoformu-

lations: a future nanomedicine for cancer. Drug Discov. Today 17, 71–80.

[38] Cheng, A. L., Hsu, C. H., Lin, J. K., Hsu, M. M., Ho, Y. F., et al. (2001) Phase

I clinical trial of curcumin, a chemopreventive agent, in patients with high-

risk or pre-malignant lesions. Anticancer Res. 21, 2895–2900.

[39] Sarkar, F. H., Li, Y., Wang, Z., and Padhye, S. (2010) Lesson learned from

nature for the development of novel anti-cancer agents: implication of iso-

flavone, curcumin, and their synthetic analogs. Curr. Pharm. Des. 16,

1801–1812.

[40] Shehzad, A., Wahid, F., and Lee, Y. S. (2010) Curcumin in cancer chemo-

prevention: molecular targets, pharmacokinetics, bioavailability, and clini-

cal trials. Arch. Pharm. 343, 489–499.

[41] Singh, R. and Sharma, P. (2011) Hepatoprotective effect of curcumin on

lindane-induced oxidative stress in male wistar rats. Toxicol. Int. 18,

124–129.

[42] Girish, C. and Pradhan, S. C. (2012) Hepatoprotective activities of

picroliv, curcumin, and ellagic acid compared to silymarin on carbon-tetra-

chloride-induced liver toxicity in mice. J. Pharmacol. Pharmacother. 3,

149–155.

[43] Morsy, M. A., Abdalla, A. M., Mahmoud, A. M., Abdelwahab, S. A., and

Mahmoud, M. E. (2012) Protective effects of curcumin, alpha-lipoic acid,

and N-acetylcysteine against carbon tetrachloride-induced liver fibrosis in

rats. J. Physiol. Biochem. 68, 29–35.

[44] Singh, M., Sasi, P., Gupta, V. H., Rai, G., Amarapurkar, D. N., et al. (2012)

Protective effect of curcumin, silymarin and N-acetylcysteine on antituber-

cular drug-induced hepatotoxicity assessed in an in vitro model. Hum.

Exp. Toxicol. 31, 788–797.

[45] Srivastava, R., Dikshit, M., Srimal, R. C., and Dhawan, B. N. (1985) Anti-

thrombotic effect of curcumin. Thromb. Res. 40, 413–417.

[46] Prakash, P., Misra, A., Surin, W. R., Jain, M., Bhatta, R. S., et al. (2011)

Anti-platelet effects of Curcuma oil in experimental models of myocardial

ischemia-reperfusion and thrombosis. Thromb. Res. 127, 111–118.

[47] Miriyala, S., Panchatcharam, M., and Rengarajulu, P. (2007) Cardioprotec-

tive effects of curcumin. Adv. Exp. Med. Biol. 595, 359–377.

[48] Duan, W., Yang, Y., Yan, J., Yu, S., Liu, J., et al. (2012) The effects of cur-

cumin post-treatment against myocardial ischemia and reperfusion by

activation of the JAK2/STAT3 signaling pathway. Basic Res. Cardiol. 107,

263.

Monroy et al. 129

Page 9: Curcumin and neurodegenerative diseases

[49] Izem-Meziane, M., Djerdjouri, B., Rimbaud, S., Caffin, F., Fortin, D., et al.

(2012) Catecholamine-induced cardiac mitochondrial dysfunction and

mPTP opening: protective effect of curcumin. Am. J. Physiol. Heart Circ.

Physiol. 302, H665–H674.

[50] Reddy, R. C., Vatsala, P. G., Keshamouni, V. G., Padmanaban, G., and Ran-

garajan, P. N. (2005) Curcumin for malaria therapy. Biochem. Biophys.

Res. Commun. 326, 472–474.

[51] Perez-Arriaga, L., Mendoza-Magana, M. L., Cortes-Zarate, R., Corona-Riv-

era, A., Bobadilla-Morales, L., et al. (2006) Cytotoxic effect of curcumin on

Giardia lamblia trophozoites. Acta Trop. 98, 152–161.

[52] De, R., Kundu, P., Swarnakar, S., Ramamurthy, T., Chowdhury, A., et al. (2009)

Antimicrobial activity of curcumin against Helicobacter pylori isolates from

India and during infections in mice. Antimicrob. Agents Chemother. 53,

1592–1597.

[53] Na, H. S., Cha, M. H., Oh, D. R., Cho, C. W., Rhee, J. H., et al. (2011) Pro-

tective mechanism of curcumin against Vibrio vulnificus infection. FEMS

Immunol. Med. Microbiol. 63, 355–362.

[54] Nagajyothi, F., Zhao, D., Weiss, L. M., and Tanowitz, H. B. (2012) Curcumin

treatment provides protection against Trypanosoma cruzi infection. Parasi-

tol. Res. 110, 2491–2499.

[55] Ye, J. and Zhang, Y. (2012) Curcumin protects against intracellular amyloid

toxicity in rat primary neurons. Int. J. Clin. Exp. Med. 5, 44–49.

[56] Lavoie, S., Chen, Y., Dalton, T. P., Gysin, R., Cu�enod, M., et al. (2009) Cur-

cumin, quercetin, and tBHQ modulate glutathione levels in astrocytes and

neurons: importance of the glutamate cysteine ligase modifier subunit. J.

Neurochem. 108, 1410–1422.

[57] Karlstetter, M., Lippe, E., Walczak, Y., Moehle, C., Aslanidis, A., et al.

(2011) Curcumin is a potent modulator of microglial gene expression and

migration. J. Neuroinflamm. 8, 125.

[58] Wang, J., Zhang, Y. J., and Du, S. (2012) The protective effect of curcumin

on Abeta induced aberrant cell cycle reentry on primary cultured rat corti-

cal neurons. Eur. Rev. Med. Pharmacol. Sci. 16, 445–454.

[59] Ortiz-Ortiz, M. A., Mor�an, J. M., Ruiz-Mesa, L. M., Niso-Santano, M., Bravo-

SanPedro, J. M., et al. (2010) Curcumin exposure induces expression of

the Parkinson’s disease-associated leucine-rich repeat kinase 2 (LRRK2) in

rat mesencephalic cells. Neurosci. Lett. 468, 120–124.

[60] Jiang, H., Tian, X., Guo, Y., Duan, W., Bu, H., et al. (2011) Activation of nu-

clear factor erythroid 2-related factor 2 cytoprotective signaling by curcu-

min protect primary spinal cord astrocytes against oxidative toxicity. Biol.

Pharm. Bull. 34, 1194–1197.

[61] Darvesh, A. S., Carroll, R. T., Bishayee, A., Novotny, N. A., Geldenhuys, W.

J., et al. (2012) Curcumin and neurodegenerative diseases: a perspective.

Expert Opin. Investig. Drugs 21, 1123–1140.

[62] Aggarwal, B. B. and Sung, B. (2009) Pharmacological basis for the role of

curcumin in chronic diseases: an age-old spice with modern targets.

Trends Pharmacol. Sci. 30, 85–94.

[63] Kelsey, N. A., Wilkins, H. M., and Linseman, D. A. (2010) Nutraceutical

antioxidants as novel neuroprotective agents. Molecules 15, 7792–

7814.

[64] Agrawal, R., Mishra, B., Tyagi, E., Nath, C., and Shukla, R. (2010) Effect of

curcumin on brain insulin receptors and memory functions in STZ (ICV)

induced dementia model of rat. Pharmacol. Res. 61, 247–252.

[65] Zhou, H., Beevers, C. S., and Huang, S. (2011) The targets of curcumin.

Curr. Drug Targets 12, 332–347.

[66] Ono, K., Hasegawa, K., Naiki, H., and Yamada, M. (2004) Curcumin has

potent anti-amyloidogenic effects for Alzheimer’s beta-amyloid fibrils in

vitro. J. Neurosci. Res. 75, 742–750.

[67] Kim, H., Park, B. S., Lee, K. G., Choi, C. Y., Jang, S. S., et al. (2005) Effects

of naturally occurring compounds on fibril formation and oxidative stress

of beta-amyloid. J. Agric. Food Chem. 53, 8537–8541.

[68] Shytle, R. D., Bickford, P. C., Rezai-zadeh, K., Hou, L., Zeng, J., et al. (2009)

Optimized turmeric extracts have potent anti-amyloidogenic effects. Curr.

Alzheimer Res. 6, 564–571.

[69] Guo, J. P., Yu, S., and McGeer, P. L. (2010) Simple in vitro assays to iden-

tify amyloid-beta aggregation blockers for Alzheimer’s disease therapy. J.

Alzheimers Dis. 19, 1359–1370.

[70] Varghese, K., Molnar, P., Das, M., Bhargava, N., Lambert, S., et al. (2010)

A new target for amyloid beta toxicity validated by standard and high-

throughput electrophysiology. PLoS One 5, e8643.

[71] Zhang, C., Browne, A., Child, D., and Tanzi, R. E. (2010) Curcumin

decreases amyloid-beta peptide levels by attenuating the maturation of

amyloid-beta precursor protein. J. Biol. Chem. 285, 28472–28480.

[72] Kuner, P., Schubenel, R., and Hertel, C. (1998) Beta-amyloid binds

to p57NTR and activates NFkappaB in human neuroblastoma cells.

J. Neurosci. Res. 54, 798–804.

[73] Kim, D. S., Park, S. Y., and Kim, J. K. (2001) Curcuminoids from Curcuma

longa L. (Zingiberaceae) that protect PC12 rat pheochromocytoma and

normal human umbilical vein endothelial cells from betaA(1–42) insult.

Neurosci. Lett. 303, 57–61.

[74] Ono, K. and Yamada, M. (2006) Antioxidant compounds have potent anti-

fibrillogenic and fibril-destabilizing effects for alpha-synuclein fibrils in

vitro. J. Neurochem. 97, 105–115.

[75] Cho, J. W., Lee, K. S., and Kim, C. W. (2007) Curcumin attenuates

the expression of IL-1beta, IL-6, and TNF-alpha as well as cyclin E in TNF-

alpha-treated HaCaT cells; NF-kappaB and MAPKs as potential upstream

targets. Int. J. Mol. Med. 19, 469–474.

[76] Cole, G. M., Teter, B., and Frautschy, S. A. (2007) Neuroprotective effects

of curcumin. Adv. Exp. Med. Biol. 595, 197–212.

[77] Rogers, J. T., Randall, J. D., Eder, P. S., Huang, X., Bush, A. I., et al. (2002)

Alzheimer’s disease drug discovery targeted to the APP mRNA

50untranslated region. J. Mol. Neurosci. 19, 77–82.

[78] Jomova, K., Vondrakova, D., Lawson, M., and Valko, M. (2010) Metals, oxi-

dative stress and neurodegenerative disorders. Mol. Cell. Biochem. 345,

91–104.

[79] Baum, L. and Ng, A. (2004) Curcumin interaction with copper and iron

suggests one possible mechanism of action in Alzheimer’s disease animal

models. J. Alzheimers Dis. 6, 367–377; discussion 443–369.

[80] Barik, A., Mishra, B., Kunwar, A., Kadam, R. M., Shen, L., et al. (2007)

Comparative study of copper(II)-curcumin complexes as superoxide dis-

mutase mimics and free radical scavengers. Eur. J. Med. Chem. 42,

431–439.

[81] Beal, M. F. (2001) Experimental models of Parkinson’s disease. Nat. Rev.

Neurosci. 2, 325–334.

[82] Wang, J., Du, X. X., Jiang, H., and Xie, J. X. (2009) Curcumin attenuates 6-

hydroxydopamine-induced cytotoxicity by anti-oxidation and nuclear fac-

tor-kappa B modulation in MES23.5 cells. Biochem. Pharmacol. 78,

178–183.

[83] Jaisin, Y., Thampithak, A., Meesarapee, B., Ratanachamnong, P., Suksam-

rarn, A., et al. (2011) Curcumin I protects the dopaminergic cell line SH-

SY5Y from 6-hydroxydopamine-induced neurotoxicity through attenuation

of p53-mediated apoptosis. Neurosci. Lett. 489, 192–196.

[84] Chen, J., Tang, X. Q., Zhi, J. L., Cui, Y., Yu, H. M., et al. (2006) Curcumin

protects PC12 cells against 1-methyl-4-phenylpyridinium ion-induced apo-

ptosis by bcl-2-mitochondria-ROS-iNOS pathway. Apoptosis 11, 943–953.

[85] Yu, S., Zheng, W., Xin, N., Chi, Z. H., Wang, N. Q., et al. (2010) Curcumin

prevents dopaminergic neuronal death through inhibition of the c-Jun N-

terminal kinase pathway. Rejuvenation Res. 13, 55–64.

[86] Novak, J. and Vanek, Z. (1992) Screening for a new generation of anthel-

minthic compounds. In vitro selection of the nematode Caenorhabditis ele-

gans for ivermectin resistance. Folia Microbiol. 37, 237–238.

[87] Davies, A. G. and McIntire, S. L. (2004) Using C. elegans to screen for tar-

gets of ethanol and behavior-altering drugs. Biol. Proced Online 6,

113–119.

[88] Kwok, T. C., Ricker, N., Fraser, R., Chan, A. W., Burns, A., et al. (2006) A

small-molecule screen in C. elegans yields a new calcium channel antago-

nist. Nature 441, 91–95.

[89] Tampakakis, E., Okoli, I., and Mylonakis, E. (2008) A C. elegans-based,

whole animal, in vivo screen for the identification of antifungal

compounds. Nat. Protoc. 3, 1925–1931.

[90] Moy, T. I., Conery, A. L., Larkins-Ford, J., Wu, G., Mazitschek, R., et al.

(2009) High-throughput screen for novel antimicrobials using a whole ani-

mal infection model. ACS Chem. Biol. 4, 527–533.

BioFactors

130 Neuroprotective Effects of Curcumin

Page 10: Curcumin and neurodegenerative diseases

[91] Lemieux, G. A., Liu, J., Mayer, N., Bainton, R. J., Ashrafi, K., et al. (2011) A

whole-organism screes identifies new regulators of fat storage. Nat.

Chem. Biol. 7, 206–213.

[92] Harrington, L. A. and Harley, C. B. (1988) Effect of vitamin E on lifespan

and reproduction in Caenorhabditis elegans. Mech. Ageing Dev. 43,

71–78.

[93] Adachi, H. and Ishii, N. (2000) Effects of tocotrienols on life span and pro-

tein carbonylation in Caenorhabditis elegans. J. Gerontol. A Biol. Sci.

Med. Sci. 55, B280–B285.

[94] Melov, S., Ravenscroft, J., Malik, S., Gill, M. S., Walker, D. W., et al. (2000)

Extension of life-span with superoxide dismutase/catalase mimetics. Sci-

ence 289, 1567–1569.

[95] Fei, Y. J., Inoue, K., and Ganapathy, V. (2003) Structural and functional

characteristics of two sodium-coupled dicarboxylate transporters

(ceNaDC1 and ceNaDC2) from Caenorhabditis elegans and their relevance

to life span. J. Biol. Chem. 278, 6136–6144.

[96] Evason, K., Huang, C., Yamben, I., Covey, D. F., and Kornfeld, K. (2005)

Anticonvulsant medications extend worm life-span. Science 307, 258–

262.

[97] Benedetti, M. G., Foster, A. L., Vantipalli, M. C., White, M. P., Sampayo, J.

N., et al. (2008) Compounds that confer thermal stress resistance and

extended lifespan. Exp. Gerontol. 43, 882–891.

[98] McColl, G., Killilea, D. W., Hubbard, A. E., Vantipalli, M. C., Melov, S., et al.

(2008) Pharmacogenetic analysis of lithium-induced delayed aging in Cae-

norhabditis elegans. J. Biol. Chem. 283, 350–357.

[99] Srivastava, D., Arya, U., SoundaraRajan, T., Dwivedi, H., Kumar, S., et al.

(2008) Reserpine can confer stress tolerance and lifespan extension in the

nematode C. elegans. Biogerontology 9, 309–316.

[100] Pietsch, K., Saul, N., Menzel, R., Sturzenbaum, S. R., and Steinberg, C. E.

(2009) Quercetin mediated lifespan extension in Caenorhabditis elegans

is modulated by age-1, daf-2, sek-1 and unc-43. Biogerontology 10,

565–578.

[101] Saul, N., Pietsch, K., Menzel, R., Sturzenbaum, S. R., and Steinberg, C. E.

(2009) Catechin induced longevity in C. elegans: from key regulator genes

to disposable soma. Mech. Ageing Dev. 130, 477–486.

[102] Honda, Y., Tanaka, M., and Honda, S. (2010) Trehalose extends longevity

in the nematode Caenorhabditis elegans. Aging Cell 9, 558–569.

[103] Onken, B. and Driscoll, M. (2010) Metformin induces a dietary restriction-

like state and the oxidative stress response to extend C. elegans Health-

span via AMPK, LKB1, and SKN-1. PLoS One 5, e8758.

[104] Alavez, S., Vantipalli, M. C., Zucker, D. J., Klang, I. M., and Lithgow, G. J.

(2011) Amyloid-binding compounds maintain protein homeostasis during

ageing and extend lifespan. Nature 472, 226–229.

[105] Powolny, A. A., Singh, S. V., Melov, S., Hubbard, A., and Fisher, A. L.

(2011) The garlic constituent diallyl trisulfide increases the lifespan of C.

elegans via skn-1 activation. Exp. Gerontol. 46, 441–452.

[106] Wu, Z., Smith, J. V., Paramasivam, V., Butko, P., Khan, I., et al. (2002)

Ginkgo biloba extract EGb 761 increases stress resistance and extends life

span of Caenorhabditis elegans. Cell. Mol. Biol. 48, 725–731.

[107] Wilson, M. A., Shukitt-Hale, B., Kalt, W., Ingram, D. K., Joseph, J. A., et al.

(2006) Blueberry polyphenols increase lifespan and thermotolerance in

Caenorhabditis elegans. Aging Cell 5, 59–68.

[108] Bass, T. M., Weinkove, D., Houthoofd, K., Gems, D., and Partridge, L.

(2007) Effects of resveratrol on lifespan in Drosophila melanogaster and

Caenorhabditis elegans. Mech. Ageing Dev. 128, 546–552.

[109] Strong, R., Miller, R. A., Astle, C. M., Baur, J. A., de Cabo, R., et al. (2012)

Evaluation of resveratrol, green tea extract, curcumin, oxaloacetic acid,

and medium-chain triglyceride oil on life span of genetically heterogene-

ous mice. J. Gerontol. A Biol. Sci. Med. Sci.

[110] Link, C. D., Taft, A., Kapulkin, V., Duke, K., Kim, S., et al. (2003) Gene

expression analysis in a transgenic Caenorhabditis elegans Alzheimer’s

disease model. Neurobiol. Aging 24, 397–413.

[111] Kraemer, B. C., Zhang, B., Leverenz, J. B., Thomas, J. H., Trojanowski, J.

Q., et al. (2003) Neurodegeneration and defective neurotransmission in a

Caenorhabditis elegans model of tauopathy. Proc. Natl. Acad. Sci. USA

100, 9980–9985.

[112] Miyasaka, T., Ding, Z., Gengyo-Ando, K., Oue, M., Yamaguchi, H., et al.

(2005) Progressive neurodegeneration in C. elegans model of tauopathy.

Neurobiol. Dis. 20, 372–383.

[113] Lakso, M., Vartiainen, S., Moilanen, A. M., Sirvio, J., Thomas, J. H., et al.

(2003) Dopaminergic neuronal loss and motor deficits in Caenorhabditis

elegans overexpressing human alpha-synuclein. J. Neurochem. 86,

165–172.

[114] Cao, S., Gelwix, C. C., Caldwell, K. A., and Caldwell, G. A. (2005) Torsin-

mediated protection from cellular stress in the dopaminergic neurons of

Caenorhabditis elegans. J. Neurosci. 25, 3801–3812.

[115] Hamamichi, S., Rivas, R. N., Knight, A. L., Cao, S., Caldwell, K. A., et al.

(2008) Hypothesis-based RNAi screening identifies neuroprotective genes

in a Parkinson’s disease model. Proc. Natl. Acad. Sci. USA 105,

728–733.

[116] van Ham, T. J., Thijssen, K. L., Breitling, R., Hofstra, R. M., Plasterk, R. H.,

et al. (2008) C. elegans model identifies genetic modifiers of alpha-synu-

clein inclusion formation during aging. PLoS Genet. 4, e1000027.

[117] Park, K. W. and Li, L. (2008) Cytoplasmic expression of mouse prion pro-

tein causes severe toxicity in Caenorhabditis elegans. Biochem. Biophys.

Res. Commun. 372, 697–702.

[118] Gidalevitz, T., Ben-Zvi, A., Ho, K. H., Brignull, H. R., and Morimoto, R. I.

(2006) Progressive disruption of cellular protein folding in models of poly-

glutamine diseases. Science 311, 1471–1474.

[119] Gidalevitz, T., Krupinski, T., Garcia, S., and Morimoto, R. I. (2009) Destabi-

lizing protein polymorphisms in the genetic background direct phenotypic

expression of mutant SOD1 toxicity. PLoS Genet. 5, e1000399.

[120] Alavez, S. and Lithgow, G. J. (2012) Pharmacological maintenance of pro-

tein homeostasis could postpone age-related disease. Aging Cell 11,

187–191.

[121] Hsu, A. L., Murphy, C. T., and Kenyon, C. (2003) Regulation of aging and

age-related disease by DAF-16 and heat-shock factor. Science 300,

1142–1145.

[122] Rincon-Limas, D. E., Jensen, K., and Fernandez-Funez, P. (2012) Drosophila

models of proteinopathies: the little fly that could. Curr. Pharm. Des. 18,

1108–1122.

[123] Caesar, I., Jonson, M., Nilsson, K. P., Thor, S., and Hammarstrom, P.

(2012) Curcumin promotes A-beta fibrillation and reduces neurotoxicity in

transgenic Drosophila. PLoS One 7, e31424.

[124] Strong, R., Miller, R. A., Astle, C. M., Floyd, R. A., Flurkey, K., et al. (2008)

Nordihydroguaiaretic acid and aspirin increase lifespan of genetically het-

erogeneous male mice. Aging Cell 7, 641–650.

[125] Harrison, D. E., Strong, R., Sharp, Z. D., Nelson, J. F., Astle, C. M., et al.

(2009) Rapamycin fed late in life extends lifespan in genetically heteroge-

neous mice. Nature 460, 392–395.

[126] Miller, R. A., Harrison, D. E., Astle, C. M., Baur, J. A., Boyd, A. R., et al.

(2011) Rapamycin, but not resveratrol or simvastatin, extends life span of

genetically heterogeneous mice. J. Gerontol. A Biol. Sci. Med. Sci. 66,

191–201.

[127] Frautschy, S. A., Hu, W., Kim, P., Miller, S. A., Chu, T., et al. (2001) Pheno-

lic anti-inflammatory antioxidant reversal of Abeta-induced cognitive defi-

cits and neuropathology. Neurobiol. Aging 22, 993–1005.

[128] Lim, G. P., Chu, T., Yang, F., Beech, W., Frautschy, S. A., et al. (2001) The

curry spice curcumin reduces oxidative damage and amyloid pathology in

an Alzheimer transgenic mouse. J. Neurosci. 21, 8370–8377.

[129] Begum, A. N., Jones, M. R., Lim, G. P., Morihara, T., Kim, P., et al. (2008)

Curcumin structure-function, bioavailability, and efficacy in models of neu-

roinflammation and Alzheimer’s disease. J. Pharmacol. Exp. Ther. 326,

196–208.

[130] Pan, R., Qiu, S., Lu, D. X., and Dong, J. (2008) Curcumin improves learning

and memory ability and its neuroprotective mechanism in mice. Chin.

Med. J. 121, 832–839.

[131] Pandey, N., Strider, J., Nolan, W. C., Yan, S. X., and Galvin, J. E. (2008)

Curcumin inhibits aggregation of alpha-synuclein. Acta Neuropathol. 115,

479–489.

[132] Zbarsky, V., Datla, K. P., Parkar, S., Rai, D. K., Aruoma, O. I., et al. (2005)

Neuroprotective properties of the natural phenolic antioxidants curcumin

Monroy et al. 131

Page 11: Curcumin and neurodegenerative diseases

and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkin-

son’s disease. Free Radic. Res. 39, 1119–1125.

[133] Tripanichkul, W. and Jaroensuppaperch, E. O. (2012) Curcumin protects ni-

grostriatal dopaminergic neurons and reduces glial activation in 6-hy-

droxydopamine hemiparkinsonian mice model. Int. J. Neurosci. 122,

263–270.

[134] Vajragupta, O., Boonchoong, P., Watanabe, H., Tohda, M., Kummasud, N.,

et al. (2003) Manganese complexes of curcumin and its derivatives: evalu-

ation for the radical scavenging ability and neuroprotective activity. Free

Radic. Biol. Med. 35, 1632–1644.

[135] Rajeswari, A. (2006) Curcumin protects mouse brain from oxidative stress

caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Eur. Rev. Med.

Pharmacol. Sci. 10, 157–161.

[136] Rajeswari, A. and Sabesan, M. (2008) Inhibition of monoamine oxidase-B

by the polyphenolic compound, curcumin and its metabolite tetrahydro-

curcumin, in a model of Parkinson’s disease induced by MPTP neurode-

generation in mice. Inflammopharmacology 16, 96–99.

[137] Mythri, R. B., Harish, G., Dubey, S. K., Misra, K., and Bharath, M. M. (2011)

Glutamoyl diester of the dietary polyphenol curcumin offers improved pro-

tection against peroxynitrite-mediated nitrosative stress and damage of

brain mitochondria in vitro: implications for Parkinson’s disease. Mol. Cell.

Biochem. 347, 135–143.

[138] Hickey, M. A., Zhu, C., Medvedeva, V., Lerner, R. P., Patassini, S., et al.

(2012) Improvement of neuropathology and transcriptional deficits in CAG

140 knock-in mice supports a beneficial effect of dietary curcumin in Hun-

tington’s disease. Mol. Neurodegener. 7, 12.

[139] Ng, T. P., Chiam, P. C., Lee, T., Chua, H. C., Lim, L., et al. (2006) Curry consump-

tion and cognitive function in the elderly. Am. J. Epidemiol. 164, 898–906.

[140] Baum, L., Lam, C. W., Cheung, S. K., Kwok, T., Lui, V., et al. (2008) Six-month

randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin

in patients with Alzheimer disease. J. Clin. Psychopharmacol. 28, 110–113.

[141] Ringman, J. M., Younkin, S. G., Pratico, D., Seltzer, W., Cole, G. M., et al.

(2008) Biochemical markers in persons with preclinical familial Alzheimer

disease. Neurology 71, 85–92.

BioFactors

132 Neuroprotective Effects of Curcumin