You are on page 1of 22

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/331405201

Antioxidant Effects of Curcumin in Models of Neurodegeneration, Ageing,


Oxidative and NITROSATIVE Stress: A Review

Article  in  Neuroscience · February 2019


DOI: 10.1016/j.neuroscience.2019.02.020

CITATIONS READS

187 981

5 authors, including:

Shameemah Abrahams William Haylett


Stellenbosch University Stellenbosch University
29 PUBLICATIONS   565 CITATIONS    14 PUBLICATIONS   689 CITATIONS   

SEE PROFILE SEE PROFILE

Glynis Johnson Soraya Bardien


Stellenbosch University Stellenbosch University
42 PUBLICATIONS   1,251 CITATIONS    122 PUBLICATIONS   2,990 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Concussion project View project

Parkinson's Disease (PD) studies View project

All content following this page was uploaded by William Haylett on 20 March 2019.

The user has requested enhancement of the downloaded file.


NEUROSCIENCE
REVIEW
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

Antioxidant effects of curcumin in models of neurodegeneration, aging,


oxidative and nitrosative stress: A review
Shameemah Abrahams,a,* William L. Haylett, a,b Glynis Johnson,c Jonathan A. Carr,d and Soraya Bardien a
a
Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University,
South Africa
b
Division of Endocrinology, Department of Medicine, Stellenbosch University, Cape Town, South Africa
c
DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division
of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
d
Division of Neurology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa

Abstract—The global burden of neurodegenerative disorders has increased substantially over the past 2 decades due to
rising rates of population aging. Although neurodegenerative disorders differ in their clinical presentation, the underlying
pathobiological processes are largely shared. Oxidative stress, among other mechanisms, is strongly implicated in
neurodegenerative disorders and aging, and can potentially be targeted by antioxidative agents. Curcumin, a component
of turmeric, is a compound that has received considerable attention for its therapeutic properties, and it is considered
to be a powerful antioxidant. In this review, we analyzed the evidence for curcumin as an antioxidant in models of neuro-
degenerative disorders as well as oxido-nitrosative stress. A total of 1451 articles were found from 3 scientific literature
databases (PubMed, Scopus, and Web of Science). After all exclusions, a final total of 64 articles were included in
this review. The majority of the studies showed that curcumin, or derivatives thereof, were protective against oxidative
and/or nitrosative stress in various cellular and animal models. Overall, curcumin protected against lipid and protein
oxidation with a reduction in levels of malondialdehyde, and protein carbonyls, thiols and nitrotyrosines. Furthermore,
it stimulated the activities of antioxidant enzymes such as superoxide dismutase and glutathione peroxidase. In

*Corresponding author.
E-mail address: shameemah@sun.ac.za (Shameemah Abrahams).
Abbreviations: 3-NP, 3-nitropropionic acid; 6-OHDA, 6-hydroxydopamine; Aβ40 or Aβ42, 40- or 42-amino acid amyloid-β peptide; A172, human glioma cells;
A53T, point-mutation resulting in alanine to threonine change at amino acid 53 of α-synuclein; AD, Alzheimer's disease; Akt, serine/threonine protein kinase
B; AKR1B4, aldo-keto reductase family 1, member B4; AKR1B8, aldo-keto reductase family 1, member B8; ALAS1, 5′-aminolevulinate synthase 1; AlCl3,
aluminum chloride; AP-1, activated protein 1; APE1, apurinic/apyrimidinic endonuclease; AOX1, aldehyde oxidase 1; APP, amyloid precursor protein;
ARE, antioxidant responsive element; b.w., body weight; C6, rat glioma cells; Ca2+, calcium; CAT, catalase; curcumin D1, di-piperoyl; CoCl2, cobalt chloride;
Cu(II), copper(II); curcumin D2, di-valinoyl; curcumin D3, di-glutamoyl; DAF-FM DA, 4-Amino-5-methylamino-2′,7′-difluorofluorescein diacetate; DCHF-DA,
dichlorodihydrofluorescein diacetate; DHR123, dihydrorhodamine 123; DMC, demethoxycurcumin; DMSO, dimethyl sulfoxide; DNPH, 2,4-dinitrophenylhydrazine;
DPPH, 2,2-diphenyl-1-picryl-hydrazyl-hydrate; DTNB, 5,5-dithio-bis-(2-nitrobenzoic acid) or Ellman's reagent; ELISA, enzyme-linked immunosorbent assay; FRAP,
ferric reducing antioxidant power; G6PD, glucose-6-phosphate dehydrogenase; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GFAP, glial fibrillary acidic
protein; GFP, green fluorescent protein; GGT1, gamma-glutamyltransferase 1; GPx, glutathione peroxidase; GR, glutathione reductase; GSH, glutathione; GST,
glutathione S-transferase; GSTA5, glutathione S-transferase alpha 5; GSTD1, glutathione S-transferase D1; GSTP1, glutathione S-transferase pi 1; h, hours;
H2O2, hydrogen peroxide; HA-sp, human spinal cord-derived astrocytes; HD, Huntington's disease; HEK293, human kidney cells; HFP, hydroxyl radical
and peroxynitrite sensor; HMOX1, heme oxygenase 1; HNE, 4-hydroxy-2-nonenal; HT22, mouse hippocampal cells; ICV-STZ, intracerebroventricular–
streptozotocin; IκBα, inhibitors-of-kappaB; IL-6, interleukin-6; IMR-32, human brain neuroblastoma cells; iNOS, inducible nitrogen oxide synthase; i.p.,
intraperitoneally; LC, liquid chromatography; LPO, malondialdehyde-thiobarbituric assay; LPS, lipopolysaccharide; LRRK2, leucine-rich repeat kinase 2;
MAPK, mitogen-activated protein kinases; MDA, malondialdehyde; MGST1, microsomal glutathione S-transferase 1; min, minutes; MnCl2, manganese
chloride; MPP+, 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MS, multiple sclerosis; MS/MS, tandem mass spectrometry;
mth, methuselah; N27, rat dopaminergic neurons; N9, mouse microglia; NDD, neurodegenerative disorder; Neuro-2a, mouse neuroblastoma cells; NF-κB, nuclear-
factor κB; NGF, nerve growth factor; NMDA, N-methyl-D-aspartate; NQO1, NAD(P)H quinone dehydrogenase 1; NO, nitric oxide; NO2, nitrogen dioxide; N2O3,
dinitrogen trioxide; NR8383, gerbil macrophages; Nrf1/2, nuclear factor-E2-related factor 1/2; O2−, superoxide anion; O2−2, peroxide; OH, hydroxyl radical;
OLN-93, rat oligodendrocytes; PC12, rat pheochromocytoma cells; PD, Parkinson's disease; Plp1, proteolipid protein 1; PN, peroxynitrate; p.o., per os/orally
ingested; ppm, parts per million; Rh-123, rhodamine 123; RNS, reactive nitrogen species; ROS, reactive oxygen species; RT-PCR, real time polymerase
chain reaction; SAMP8, senescence accelerated mouse-prone 8; SAMR1, senescence-resistant 1 mice; s.c., subcutaneous injection; SH-SY5Y and SK-N-SH,
human bone marrow neuroblastoma cells; SMA, spinal muscular atrophy; SMN, survival of motor neuron; SOD, superoxide dismutase; SOD1/2, superoxide
dismutase 1/2; TAC, total antioxidant capacity; TBA, thiobarbituric acid; TBARS, thiobarbituric acid-reactive substances; t-BHP, tert-Butyl hydroperoxide;
TEAC, trolox equivalent antioxidant capacity assay; TNF-α, tumor necrosis factor α; TRPM2, transient receptor potential cation subfamily M, member 2, channel;
TTCA, trichloroacetic acid assay; WST, cell proliferation assay; w/v, percent weight/volume; w/w, percent mass.

https://doi.org/10.1016/j.neuroscience.2019.02.020
0306-4522/© 2019 IBRO. Published by Elsevier Ltd. All rights reserved.

1
2 Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

conclusion, curcumin appears to be a promising compound for phytomedicine. However, due to some concerns about its
efficacy, further targeted experiments are needed to identify its exact molecular targets and pathways responsible for its
antioxidant effects. © 2019 IBRO. Published by Elsevier Ltd. All rights reserved.

Key words: curcumin, neurodegenerative disorders, antioxidant, oxidative stress, nitrosative stress, curcumin derivatives.

INTRODUCTION compared to free curcumin (Jangra et al., 2016; Shelat and


Acharya, 2016). However, other studies showed that curcu-
Neurodegenerative disorders (NDDs) are becoming significant
min had a greater protective effect compared to the deriva-
global health burdens which are exacerbated by escalating
tives; while altering curcumin's structure reduced the
numbers of the elderly in both developed and developing
efficacy as an antioxidant (Begum et al., 2008; Agrawal
countries (Kassebaum et al., 2016). Most NDDs, including
et al., 2012). Hence, curcumin may be a beneficial anti-
Alzheimer's disease (AD), Parkinson's disease (PD) and
oxidative and neuroprotective agent with valuable clinical
Huntington's disease (HD) remain essentially incurable. The
implications for NDDs if the technical challenges can be
paucity of therapeutic options to prevent or treat NDDs is of
met. Here, we review the evidence of the antioxidant proper-
concern, as the prevalence and disability resulting from these
ties of curcumin in various models of NDDs, aging as well
disorders are predicted to increase significantly (WHO, 2006).
as oxidative and nitrosative stress.
NDDs are characterized by degenerative processes and are
thought to share several pathobiological pathways. Inflamma-
tion, impaired protein processing, mitochondrial dysfunction,
oxidative stress, and disruptions in the autophagy-lysosomal
OXIDO-NITROSATIVE STRESS PATHWAYS
system (Vidoni et al., 2016; Wolfe, 2018) are proposed as A salient feature of NDDs is the presence of oxido-nitrosative
pathobiological pathways where initial perturbations in 1 stress, or the imbalance of reactive oxygen and nitrogen
pathway may cascade into a vicious cycle of cellular impair- species (ROS and RNS), in affected neurons. In order to
ment and neurodegeneration. Of particular interest is the contextualize the experimental findings of the reviewed litera-
oxidative stress response, which is strongly implicated in ture, we provide a brief introduction to the ROS and RNS
NDDs and aging, and which can potentially be targeted by pathways and how these are experimentally measured. In
antioxidative agents (Jiang et al., 2016; Liguori et al., 2018). cells, free radicals are unstable compounds which readily bind
One antioxidative compound that has received considerable to oxygen to become reactive species such as ROS and
attention is the natural polyphenol curcumin, one of the RNS, with many cytotoxic consequences (Gebicki, 2016).
major components of turmeric. A growing body of evidence Free radicals can form in response to external factors includ-
reports the various antioxidative, anticarcinogenic and anti- ing exposure to radiation, environmental pollution or pesti-
inflammatory effects of curcumin across many cellular and cides, as well as, due to internal bodily processes such as
animal models of different disorders (Pulido-Moran et al., energy metabolism, phagocytosis and inflammation (Lobo
2016), while chemical modification of this promising com- et al., 2010). Physiological levels of ROS and RNS also play
pound can increase its bioavailability, structural stability and important roles in cell signaling and the innate immune
efficacy (Oliveira et al., 2015). While curcumin potentially system; hence, an imbalance in ROS and RNS levels can
has many protective properties, the poor intestinal absorp- shift the scale towards being either damaging or beneficial.
tion, structural instability, limited blood–brain barrier penetra- Common cellular free radicals include superoxide anion
tion and rapid degradation of curcumin in the body limit the (O2−), hydrogen peroxide (H2O2), hydroxyl radical (OH) and
potential as a therapeutic agent in clinical trials (Ullah et al., nitric oxide (NO) (Finkel and Holbrook, 2000; Bharath et al.,
2017). Studies reported lower plasma curcumin compared 2002; Hayyan et al., 2016). Cellular O2− is highly reactive
to the ingested amount of curcumin, for example one study and is reduced by superoxide dismutase (SOD) to the less
showed that of the 8 g/day of curcumin ingested orally, only reactive H2O2, which is further reduced by catalase to water
22–41 ng/ml of curcumin was detected in the plasma (Dhillon and oxygen. In addition, ROS are neutralized by glutathione
et al., 2008; Ullah et al., 2017). The hydrophobic nature of peroxidase (GPx) in a reaction that converts the cellular
curcumin reduces its ability to solubilize in aqueous solutions antioxidant glutathione from its reduced state (GSH) to its
which poses a challenge for improving its bioavailability. oxidized form (GSSG). In turn, glutathione reductase (GR)
Therefore, studies are investigating the effect of modifying regenerates GSH from GSSG thereby replenishing the
the curcumin structure to improve bioavailability without GSH pool. Therefore, the measurement of the levels and/or
compromising on its protective properties. Studies have enzymatic activity of SOD, catalase, GPx, GSH, GSSG and
attempted to improve bioavailability by altering curcumin's GR is a useful indicator of cellular oxidative stress.
chemical structure, conjugating curcumin with lipids, encap- Cellular RNS originate from NO produced by various nitric
sulating curcumin in a nanoparticle, constructing complexes oxide synthase (NOS) enzymes, such as inducible NOS
with manganese or co-treating curcumin with piperine (iNOS), endothelial NOS and neuronal NOS. NO readily
(Chen et al., 2011, 2018; Sandhir et al., 2014; Squillaro reacts with O2− to form peroxynitrate, a slow-acting RNS
et al., 2018). In some instances, the modified curcumin which can damage cellular biomolecules. Moreover, peroxy-
has higher antioxidant ability, solubility or absorption rates nitrate can react with other molecules to form additional
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 3

RNS, including nitrogen dioxide (NO2) and dinitrogen trioxide was shown to be protective across a wide range of different
(N2O3) (Weidinger and Kozlov, 2015). concentrations and exposure times as well as for various tox-
ROS and RNS cause DNA damage, lipid peroxidation and ins applied. Further detail on each study is provided in the
protein oxidation, which damage cellular components and sections that follow.
thereby impair cellular function. The oxidation of proteins
results in the formation of protein carbonyls, thiols and nitro-
Alzheimer's disease
tyrosines, which negatively affect protein function. These
AD is the most common NDD, with an incidence of 5.3 million
compounds form readily when ROS or RNS interact with pro-
in the United States (Alzheimer Association, 2015). It is
teins and are relatively stable, which make them suitable as
characterized by widespread, progressive degeneration
markers of oxidative or nitrosative stress (Suzuki et al.,
in the brain, particularly affecting cholinergic neurons, with
2010). Similarly, peroxidation of cellular lipids by ROS and
the pathological hallmark being amyloid-β and tau fibrillary
RNS produces malondialdehyde (MDA), isoprostanes and
tangles (Braak et al., 1999).
4-hydroxy-2-nonenal (HNE), and therefore high levels of
Four studies investigated the antioxidant properties of
these are good indicators of oxidative or nitrosative stress.
curcumin in cellular models of AD (Table 1). To induce fea-
tures of AD, 2 of these involved exposure to amyloid-β, 1
LITERATURE SEARCH STRATEGY used nerve growth factor starvation and the fourth used
H2O2 exposure. The 1 study on toxic amyloid-β exposure
For this review, 1451 articles were found after literature pre-treated primary rat hippocampal neurons with the curcu-
searches in 3 databases (PubMed, Scopus, and Web of min metabolite, tetrahydrocurcumin (Mishra et al., 2011).
Science) using the following search terms: [(“curcumin”) AND Tetrahydrocurcumin reduced ROS levels when compared
(“anti-oxidant” OR “antioxidant”) AND (“neurodegenerative to the untreated and amyloid-β only treatment groups. The
disease” OR “neurodegeneration”)]. Literature searches and authors suggested that the antioxidant effects of tetrahydro-
article selection were completed during August–September curcumin protected against apoptosis. Since exposure to
2017 by 2 of the authors, independently. All non-English pesticides and environmental toxins has been implicated in
articles, non-original research articles, reviews and conference AD progression, Sarkar et al. (2017) investigated the anti-
abstracts were excluded. oxidant effects of curcumin against toxic exposure to the pes-
From our search, we selected 64 original articles that ticide, monocrotophos, in human SH-SY5Y neuroblastoma
investigated curcumin's role as an antioxidant in models of cells and amyloid-β exposure in human IMR-32 neuroblas-
neurodegeneration, aging and oxido-nitrosative stress. These toma cells. Curcumin pre-treatment reduced amyloid-β- and
included the ‘classical’ NDDs (AD, PD and HD) as well as monocrotophos-induced ROS production by 10% and 18%,
related disorders, multiple sclerosis (MS) and spinal muscular respectively. From immunofluorescence imaging, the authors
atrophy (SMA). We also included articles on dementia as it proposed that the antioxidant effect of curcumin was mediated
is considered to be relevant to the topic. These articles via the nuclear factor erythroid 2–related factor 2 (Nrf2) and
are reviewed in detail in the sections that follow and are apurinic/apyrimidinic endonuclease 1 (APE1) pathways.
summarized in Tables 1–4. The data from cellular models will In another study, nerve growth factor (NGF) receptor was
be presented first followed by animal models, with articles overexpressed in rat PC12 pheochromocytoma cells and
sub-categorized into models of (1) neurodegenerative dis- pre-treated with a cocktail of polyphenols, including curcumin
orders and (2) oxido-nitrosative stress. Following on this, (Amara et al., 2015). Subsequently, cells were exposed to
the remaining articles on dementia and aging are reported NGF-free media to elicit an AD phenotype. NGF is beneficial
on. We conclude with a section on the possible mechanistic for growth and maintenance of cholinergic neurons which
actions of curcumin as collated from the reviewed studies. makes it an important therapeutic target for AD (Ruberti
et al., 2000). ROS levels were decreased in NGF-deprived
cells pre-treated with curcumin alone or with a cocktail of poly-
STUDIES PERFORMED IN CELLULAR MODELS
phenols, including curcumin (Amara et al., 2015). In another
Antioxidant effects of curcumin in neurodegenerative study in which both mouse Neuro-2a neuroblastoma and
disorders human HEK293 kidney cells were used, curcumin was given
as a pre-treatment followed by H2O2 exposure (Morales et al.,
To date, 17 studies have investigated the antioxidant properties
2017). Curcumin protected against cell damage mediated by
of curcumin in various cellular models of NDDs (Table 1). The
oxidative stress in these cells.
majority of these studies were performed on the ‘classical’
NDDs (11 were on PD and 4 were on AD) and there was 1
each on the related disorders (SMA and MS). All of the Parkinson's disease
studies, except 2 (Van Meeteren et al., 2004; Ortiz-Ortiz PD is caused by the loss of dopaminergic neurons in the sub-
et al., 2009), showed curcumin to be protective against oxida- stantia nigra which results in a variety of motor and non-motor
tive and/or nitrosative stress. Notably, this effect was symptoms (Lees et al., 2009). The majority of PD cases have
observed irrespective of whether the curcumin treatment an unknown cause, while approximately 5–10% of cases are
was before, during or after the toxin/transfection exposure, caused by genetic mutations within genes involved in various
suggesting that curcumin's antioxidant effect is independent processes including mitochondrial health (PRKN, PINK1,
of the timing of when oxidative stress occurs. Also, curcumin LRRK2 and DJ1; Dodson and Guo, 2007).
4
Table 1. Summary of the studies investigating the effect of curcumin in cellular models of neurodegenerative disorders.

Disease Cell model Toxin applied Curcumin treatment Assay/technique Main findings References
AD Primary rat hippocampal cells Amyloid-β Curcumin derivative, tetrahydro- DCHF-DA fluorescence assay Tetrahydrocurcumin, a curcumin (Mishra et al., 2011)
(n = 9) (concentration: 2 μM; curcumin (pre-treatment concen- metabolite, reduced ROS levels
duration: 20 h) tration: 5 μM; duration: 1.5 h) induced by amyloid-β toxicity.
AD PC12 cells Nerve growth factor starvation Free CurcuminA Flow cytometry with DCHF-DA Curcumin decreased ROS levels and (Amara et al., 2015)
(rat pheochromocytoma, n = 3) (nerve growth factor-free media; (pre-treatment: 1 and 10 μM; fluorescence assay also protected against reduced mito-

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


6 or 24 h) overnight) chondrial membrane potential and cell
death.
AD Neuro-2a H2O2 Free Curcumin Immunocytochemistry, confocal Curcumin protected against cell (Morales et al., 2017)
(mouse neuroblastoma, n = 2) (50 μM; 24 h) (pre-treatment: 5 and 15 μM; 24 h) microscopy morphological damage mediated by
and HEK293 cells (human H2O2-induced oxidative stress.
kidney, n = 2)
AD IMR-32 and SH-SY5Y cells Amyloid-β and organophosphate Free Curcumin Nitroblue tetrazolium (NBT) Curcumin reduced ROS production (Sarkar et al., 2017)
(human neuroblastoma, n = 3) pesticides, monocrotophos and (pre-treatment: 10 μM; 4 h) assay, western blotting, immuno- via regulating the colocalization of
hlorpyrifos fluorescence and confocal Nrf2 and APE1.
(10 μM; 24 or 48 h) microscopy
PD SK-N-SH cells Rotenone Curcumin derivative, CNB-001 DCHF-DA fluorescence assay The neuroprotective effects of the (Jayaraj and Tamilselvam,
(human neuroblastoma, n = 4) (100 nM; 24 h) (pre-treatment: 2 μM; 2 h) curcumin derivative, CNB-001, acted 2013)
via antioxidant pathways which
assisted in mitochondrial membrane
protection and antiapoptotic effects.
PD SH-SY5Y cells Rotenone Curcumin derivative, demethoxy- DCHF-DA fluorescence assay Demethoxycurcumin, a curcumin deri- (Ramkumar et al., 2017)
(n = 4) (100 nM; 24 h) curcumin (pre-treatment: 50 nM; vative, protected against rotenone
2 h) toxicity by decreasing ROS levels.
PD SH-SY5Y cells Paraquat Free Curcumin DCHF-DA fluorescence assay, Curcumin reduced ROS levels and (Jaroonwitchawan et al.,
(n = 3) (0.5 mM; 24 h) (pre-treatment: 5 μM; 24 h) RT-PCR increased expression of the anti- 2017)
oxidant genes, SOD and GPX.
PD Transfected SH-SY5Y cells Transfected wild type Free Curcumin DCHF-DA fluorescence assay Curcumin suppressed ROS produc- (Wang et al., 2010)
(n = 3) α-synuclein (using (48 h post-transfection: tion induced by both oligomeric and
α-synuclein/GFP fusion 4 μM; 24 h) intracellular α-synuclein
protein plasmid DNA) aggregates.
PD PC12 cells Induced mutant A53T Free Curcumin DCHF-DA fluorescence assay Curcumin reduced ROS levels and (Liu et al., 2011)
(n = 3) α-synuclein expression (co-treatment: 500 nM; protected cells against oxidative
(withdrawal of doxycycline 1–6 days) stress and cell death induced by
using tetracycline-off gene α-synuclein.
regulatory system)
PD N27 cells Buthionine sulfoximine toxin Free Curcumin and bioconjugates, LPO assay, DCHF-DA Curcumin and the bioconjugates (Harish et al., 2010)
(rat dopaminergic neurons, induced glutathione depletion curcumin D1, D2 and D3 fluorescence assay and protected against oxidative stress
n = not specified) (1.5 μM; 24 h) (pre- and post-treatments: 0.5 μM; in silicoB analyses (predictive by reducing lipid peroxidation, H2O2
24 h) GSH-depletion model) levels and ROS levels, and by also
increasing levels of GSH and GST.
Disease Cell model Toxin applied Curcumin treatment Assay/technique Main findings
PD Primary rat mesencephalic MPP +
Free Curcumin DCHF-DA fluorescence and Curcumin reduced ROS production, (Yu et al., 2016)
astrocytes (800 μM; 48 h) (pre-treatment: 8 μM; 30 min) GSH assays TNF-α and IL-6 but increased GSH
(n = not specified) levels, thereby effectively reversing
the oxidative effects of MPP+.
PD PC12 cells MPP+ Free Curcumin Flow cytometry with DHR123 Curcumin attenuated nitric oxide (Chen et al., 2006)
(n = 5) (0.5 mmol/L; 12 or 24 h) (co-treatment: 20 μmol/L; fluorescence assay synthase and ROS levels, and may
12 or 24 h) act via the glutathione- and mito-
chondria-dependent nitric oxide-
ROS pathways.
PD PC12 cells MPP+ Free Curcumin and derivatives, DPPH and TEAC assays Curcumin and curcumin derivatives (Marchiani et al., 2013)
(n = not specified) (0.5 mM; 24 h), H2O2 (75 μM; dehydrozingerone, O-methyl protected against H2O2-induced toxi-
24 h) and MnCl2 (0.75 mM; derivative, zingerone, and city. Also, curcumin had a high activity
24 h) biphenyl analogues for scavenging free radicals, which
(pre-treatment: 40 μM; 20 min) was shown by the reduced DPPH
levels and increased TEAC activity

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


(cell-free).
PD N27 cells Peroxynitrate Free Curcumin Western blotting of nitrosative The peroxynitrate levels in the mito- (Mythri et al., 2011)
(n = 3) (750 μM; 24 h), MPP+ (pre-treatment: 0.5 μM; 24 h) stress related proteins and chondria were significantly reduced
(100 μM; 24 h) biochemicalC analysis by curcumin and curcumin D3 bio-
conjugate, thereby reducing nitrosa-
tive stress.
PD N27 cells Paraquat Free Curcumin Flow cytometry, western Curcumin increased ROS production; (Ortiz-Ortiz et al., 2009)
(n = 3) (250 μM; 24 h) (co-treatment: 0.001, 0.01, blotting, TaqMan gene and however, nitrosative stress (iNOS,
0.1 μM, 10 nM; 24 h) RNA expression assay nitrotyrosine) markers were reduced.
SMA Patient-derived dermal Loss of function mutation Free Curcumin DCHF-DA fluorescence assay, Curcumin inhibited oxidative stress, (Köstel et al., 2012)
fibroblast cells in SMN gene (only treatment: 20 μM; 24 h) RT-PCR of 84 antioxidant via reducing ROS production.
(n = not specified) response genes
MS OLN-93 LPS Free Curcumin WST assay, Griess assay, Curcumin did not protect OLN-93 (Van Meeteren et al., 2004)
(rat oligodendrocytes, (1 mg/mL; single dose), (co-treatment: 2.5–25 μg/ml; western blotting of iNOS protein cells against H2O2-induced oxidative
n = 4) and NR8383 cells H2O2 (10−5–10−2 M; 3 h) 48 h) damage and LPS-induced nitrosa-
(gerbil macrophages, n = 4) tive stress.
A53T: point-mutation resulting in alanine to threonine change at amino acid 53 of α-synuclein; AD: Alzheimer's disease; APE1: apurinic/apyrimidinic endonuclease; CAT: catalase; curcumin D1: di-piperoyl; curcumin D2: di-valinoyl; curcumin D3: di-glutamoyl;
DCHF-DA: dichlorodihydrofluorescein diacetate; DPPH: 2,2-diphenyl-1-picryl-hydrazyl-hydrate; GFP: green fluorescent protein; GPX: glutathione peroxidase; GSH: glutathione; GST: glutathione S-transferase; GSTD1: glutathione S-transferase D1; h: hours;
IL-6: interleukin-6; IMR-32: human brain neuroblastoma cells; iNOS: inducible nitrogen oxide synthase; i.p.: intraperitoneally; H2O2: hydrogen peroxide; HEK293: human kidney cells; LPO: malondialdehyde-thiobarbituric assay; LPS: lipopolysaccharide,
MDA: malondialdehyde; min: minutes; MnCl2: manganese chloride; MPP+: 1-methyl-4-phenylpyridinium; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MS: multiple sclerosis; MTH: methuselah; N27: rat dopaminergic neurons; Neuro-2a:
mouse neuroblastoma cells; NGF: nerve growth factor; NR8383: gerbil macrophages; Nrf2: nuclear factor erythroid 2–related factor 2; OLN-93: rat oligodendrocytes; PC12: rat pheochromocytoma cells; PD: Parkinson's disease; p.o.: per os/orally
ingested; ROS: reactive oxygen species; RT-PCR: real time polymerase chain reaction; s.c.: subcutaneous injection; SH-SY5Y and SK-N-SH: human bone marrow neuroblastoma cells; SMA: spinal muscular atrophy; SMN: survival of motor neuron;
SOD1/2: superoxide dismutase 1/2; TBARS: thiobarbituric acid-reactive substances; TEAC: trolox equivalent antioxidant capacity assay; TNF-α: tumor necrosis factor α; WST: cell proliferation assay.
A
The antioxidant cocktail mixture contained curcumin, resveratrol, quercetin, N-acetylcysteine, hydroxytyrosol and lycopene.
B
in silico: theoretically analyzing the antioxidant effects of structural changes in curcumin using biologically-derived mathematical and computational modelling (systems biology).
C
Biochemical: analyzing the antioxidant effects of curcumin using biochemical, cell-free experiments.

5
6
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21
Table 2. Summary of the studies investigating the effect of curcumin in cellular models of oxidative stress.

Cell model Toxin applied Curcumin treatment Assay/technique Main findings References
A172 H2O2 (concentration: Free Curcumin Western blotting and Curcumin protected cells against cytotoxic (Daverey and Agrawal,
(human glioma) and HA-sp cells 50 μM; duration: 24 h) (pre- and post-treatment concentra- immunofluorescence effects of oxidative stress, including disrup- 2016)
(human spinal cord-derived astro- tions:1 μM; duration: 24 h) tion of cytonemes and F-actin aggregates,
cytes, n = not specified) decreased GFAP, vimentin, and caspase 1.
PC12 cells H2O2 Free Curcumin DCHF-DA fluorescence assay Curcumin prevented DNA damage and (Fu et al., 2016)
(rat pheochromocytoma, n = 3) (0.5 mM; 2 h) (pre-treatment: 50 and 100 μM; 12 h) oxidative stress via ROS inhibition and the
protective action of curcumin seemed to be
via the cell growth-death MAPK and Akt
pathways.
Primary astrocytes derived from H2O2 Free Curcumin DCHF-DA fluorescence assay Curcumin inhibited oxidative stress via the (Jiang et al., 2011)
knock-out mouse model (200 μM; 30 min) (pre-treatment: 10 μM; 24 h) Nrf2/ARE pathway.
(n = 3)
Neuro-2a cells H2O2 Free Curcumin Flow cytometry with DCHF-DA Curcumin reduced intracellular ROS and pro- (Zhao et al., 2011)
(mouse neuroblastoma, n = 3) (30 μM; 24 h) (pre-treatment: 20 μg/ml; 1 h) fluorescence assay tected cells against H2O2-induced damage.
Also, curcumin inhibited oxidative stress-
inducing IκBα degradation and NF-κB
nuclear translocation.
Transfected SH-SY5Y cells H2O2-induced TRPM2 Free Curcumin Rh-123 fluorescence assay Curcumin decreased ROS levels and thereby (Öz and Çelik, 2016)
(human neuroblastoma, n = 3) activation (transfected (co-treatment: 5 μM; 24 h) inhibited oxidation via TRPM2 channels.
with TRPM2)
N9 cells LPS Free Curcumin and curcumin deriva- Nitrite assay (absorbance), Curcumin and a curcumin derivative de- (Zhang et al., 2010)
(mouse microglia, n = not (1 μg/ml; 48 h) tive, DMC DCHF-DA fluorescence assay creased levels of ROS, nitrite and iNOS via
specified) (pre-treatment: 0.1–10 μM; 48 h) regulating the NF-κβ and MAPK pathways.
Primary rat cortical neurons Cu(II) Free Curcumin DCHF-DA fluorescence and Curcumin reduced oxidative ROS and H2O2 (Huang et al., 2011)
(n = not specified) (20 μM; 1 h) (pre-treatment: 1–20 μM; 23 or 24 h) hydrogen peroxide assays levels, and protected cell viability against Cu
(II)-induced oxidative damage.
Neuro-2a Iron, Fe2+ Free Curcumin Immunocytochemistry, confocal Curcumin protected against cell morphologi- (Morales et al., 2017)
(n = 2) and HEK293 cells (human (80 μM; 24 h) (pre-treatment: 5 and 15 μM; 24 h) microscopy cal damage mediated by redox iron-induced
kidney, n = 2) oxidative stress.
Cell model Toxin applied Curcumin treatment Assay/technique Main findings
Primary rat cerebellar granule Iodoacetate Free Curcumin DCHF-DA fluorescence assay Curcumin reduced iodoacetate-induced ROS (Reyes-Fermin et al., 2012)
neurons (18 μM; 30 min) (pre-treatment: 10 μM; 24 h) levels and increased heme oxygenase
(n = 4) expression, thereby inhibiting oxidative stress.
HT22 cells Hypoxia Free Curcumin DCHF-DA fluorescence assay Curcumin reduced ROS levels and thereby (Chhunchha et al., 2013)
(mouse hippocampal, n = 3) (1% O2 and CoCl2: 100 (pre-treatment: 2 μM; 12 h) regulated cellular signaling through attenua-
and 200 μM; 24–72 h) tion of hypoxia-induced oxidative and endo-
plasmic reticulum stressors.
PC12 cells Glutamate Free Curcumin DCHF-DA fluorescence assay Curcumin reduced release of Ca 2+ influx, (Chang et al., 2014)
(n = 6) (0–20 mM; 24–48 h) (pre-treatment: 5 μM; 48 h) and GSH and ROS levels which led to
reduced cell death.
Primary rat cerebellar granule Glutamate Curcumin derivative, J147 DCHF-DA, HFP, DAF-FM DA, The curcumin derivative, J147, protected (Chen et al., 2015)
and cortical neurons (75 μM; 2–24 h) (pre-treatment: 1–10 μM; 24 h) and DHR123 fluorescence against elevated levels of ROS and RNS
(n = not specified), and PC12 assays (H2O2, hydroxyl radical, nitric oxide, and
cells (n = not specified) peroxynitrite). J147 inhibited oxidative
stress induced by glutamate and t-BHP.

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


Primary rat cortical neurons and NMDA Free Curcumin RT-PCR of the iNOS gene Curcumin suppressed iNOS expression in (Lin et al., 2011)
astrocytes (n = 3) (50 μM; 30 min) (pre-treatment: 1 μM; 1 h) astrocytes under non-stressed conditions.
C6 cells none Free Curcumin Affymetrix Rat GeneChip Curcumin upregulated expression of genes (Panchal et al., 2008)
(rat glioma, n = 2) (only treatment: 5 μM; 0–48 h) expression array involved in the antioxidant response, including
the ALAS1, AKR1B8, AKR1B4, AOX1,
G6PD, GGT1, GSTA5, GSTP1, HMOX1,
MGST1, NQO1 genes.
A172: human glioma cells; Akt: serine/threonine protein kinase B; AKR1B4: aldo-keto reductase family 1, member B4; AKR1B8: aldo-keto reductase family 1, member B8; ALAS1: 5′-aminolevulinate synthase 1; AOX1: aldehyde oxidase 1;
ARE: antioxidant responsive element; C6: rat glioma cells; Ca2+: calcium; CoCl2: cobalt chloride; Cu(II): copper(II); DCHF-DA: dichlorodihydrofluorescein diacetate; DAF-FM DA: 4-Amino-5-methylamino-2′,7′-difluorofluorescein diacetate; DHR123:
dihydrorhodamine 123; DMC: demethoxycurcumin; G6PD: glucose-6-phosphate dehydrogenase; GFAP: glial fibrillary acidic protein; GGT1: gamma-glutamyltransferase 1; GSH: glutathione; GSTA5: glutathione S-transferase alpha 5; GSTP1:
glutathione S-transferase pi 1; h: hours; IκBα: inhibitors-of-kappaB; iNOS: inducible nitrogen oxide synthase; H2O2: hydrogen peroxide; HA-sp: human spinal cord-derived astrocytes; HEK293: human kidney cells; HFP: hydroxyl radical and peroxynitrite
sensor; HMOX1: heme oxygenase 1; HT22: mouse hippocampal cells; LPS: lipopolysaccharide; MAPK: mitogen-activated protein kinases; MGST1: microsomal glutathione S-transferase 1; min: minutes; N9: mouse microglia; Neuro-2a: mouse
neuroblastoma cells; NF-κB: nuclear-factor κB; NMDA: N-methyl-D-aspartate; NQO1: NAD(P)H quinone dehydrogenase 1; Nrf2: nuclear-factor -E2-related factor 2; PC12: rat pheochromocytoma cells; Rh-123: rhodamine 123; ROS: reactive oxygen
species; RNS: reactive nitrogen species; RT-PCR: real time polymerase chain reaction; SH-SY5Y: human bone marrow neuroblastoma cells; t-BHP: tert-Butyl hydroperoxide; TRPM2: transient receptor potential cation subfamily M, member 2,
channel.

7
8
Table 3. Summary of the studies investigating the effect of curcumin in animal models for neurodegenerative disorders.

Disease Animal model Toxin applied Curcumin treatment Assay/technique Main findings References
AD Transgenic male and female mice Tg2576 APPSw transgenic Free Curcumin Oxyblot kit (DNPH) Low and high-dose curcumin reduced (Lim et al.,
(n = 28; age: 10 months; low and high (overexpressing a mutant form (post-transfection concentration: 160 levels of oxidized protein carbonyls. 2001)
dose curcumin, untreated transgenic, of human APP: K670 M/N671 L) and 5000 ppm; duration: 6 months)
and untreated non-transgenic groups)
AD Sprague–Dawley female rats Aβ40 and Aβ42 Free Curcumin 8EPI-F2 immunoassay Curcumin protected against fatty acid (Frautschy
(n = 20; age: 9-month-old ‘middle-aged’ (intraventricular injection, Aβ42: (pre-treatment: 500 and 2000 ppm; kit (ELISA) oxidation by reducing isoprostane et al., 2001)
and 19-month-old ‘aged’ rats; middle- 0.2 μg, 1 μg, Aβ40: 0.7 μg; 2 months) levels (marker of lipid peroxidation).
aged: low-dose curcumin, and control 1–3 months)
groups; aged: high-dose curcumin, con-
trol, and ibuprofen groups)
AD Transgenic C57BL6/J male and female Tg2576 APPsw transgenic Free Curcumin and derivative, LC/tandem mass Curcumin decreased protein nitrotyro- (Begum et al.,
mice (overexpressing a mutant form tetrahydrocurcumin spectrometry (MS/MS) sines and carbonyls, and isoprostanes. 2008)

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


(n = 5–11; age: 3–4 months; 5 groups: of human APP: K670M/N671L) (post-transfection: 0.2–0.4 μmol or
acute and chronic treatment of curcumin and LPS-injection (100 nM; 24 h) 1.25 mg/day; 2 days or 4 months,
and tetrahydrocurcumin in both trans- for LPS-injection; pre-treatment:
genic, and non-transgenic groups) 2.5 μM; 1 h)
AD Transgenic C57BL6 × SJL F1 male and Tg2576 APP transgenic Free Curcumin Immunohistochemistry Curcumin suppressed lipid peroxida- (Seo et al.,
female mice (overexpressing a mutant form (post-transfection: 500 ppm; (HNE staining) tion in the spinal cord of APP trans- 2010)
(n = 4; age: 10 months; non-transgenic of human APP: K670M/N671L) 6 months) genic mice and rescued motor
untreated, transgenic untreated, and function deficits.
transgenic and curcumin-treated groups)
AD Transgenic Sprague–Dawley male rats Tg2576 APP transgenic Curcumin derivative, J147 Immunohistochemistry Curcumin derivative, J147, prevented (Chen et al.,
(n = 24; age: 3 months; transgenic (overexpressing a mutant form (post-transfection: 200 ppm; (HNE staining), the increase in HO-1 levels in the 2011)
untreated, non-transgenic untreated, of human APP: K670M/N671L) 5 months) spectrophotometry hippocampus.
transgenic and curcumin-treated, and (MDA levels)
non-transgenic and curcumin-treated
groups)
AD Wistar male and female rats AlCl3 Curcumin derivative, DTNB absorbance and Curcumin derivative, CUR-CA- (Shelat and
(n = 64; age: 1 month; untreated, (17 mg/kg; 15 days) CUR-CA-THIONE TBA assays THIONE suppressed the increase Acharya, 2016)
vehicle control, AlCl3-treated, curcumin (post-treatment: 500 mg/kg; in MDA levels, inhibiting lipid
control, and various curcuminoid-treat- 15 days) peroxidation, and increased GSH
ment groups) and CAT levels. CUR-CA-THIONE
also improved exploratory behavior
and locomotor function.
PD Wistar albino male rats 6-OHDA Free Curcumin and curcumin TBARS assay (lipid peroxi- Curcumin, demethoxycurcumin and (Agrawal et al.,
(n = 40; age: not specified; sham control, (10 μg/2 μL, single injection derivatives, demethoxycurcumin dation), spectrophotometer bisdemethoxycurcumin suppressed 2012)
lesion control, lesion and curcumin- in striatum) and bisdemethoxycurcumin (enzymatic activity) the 6-OHDA-induced decrease in
treated, lesion and demethoxycurcumin- (pre-treatment: 60 mg/kg orally; GSH, GPx, GR, SOD and CAT
treated, and lesion and bisdemethoxycur- 3 weeks) levels; as well as decreasing lipid
cumin-treated groups) peroxidation. Curcuminoids also
improved motor function.
PD C57BL/6 male mice MPTP Curcumin derivative, CNB-001 TBARS assay, absorbance CNB-001, the curcumin derivative, (Jayaraj et al.,
(n = 24; age: adult; vehicle control, (30 mg/kg; 4 days) (pre-treatment: 4 mg/kg; 1 h) (GSH and GPx), nitro blue inhibited oxidative stress by decreas- 2014)
MPTP only, CNB-001 only, and CNB- tetrazolium absorbance ing MDA, SOD and CAT levels while
001- and MPTP-treated groups) (SOD activity), H2O2 increasing GSH and GPx levels.
decomposition levels CNB-001 also improved locomotive
(CAT activity) and exploratory behavior
Disease Animal model Toxin applied Curcumin treatment Assay/technique Main findings
PD Lewis male rats Rotenone Free Curcumin DCHF-DA fluorescence
Curcumin suppressed the rotenone- (Cui et al.,
(n = 24; age: 7–8 weeks; vehicle con- (1 ml/kg/day, 2 injections (pre- and co-treatment: 100 mg/kg, mediated oxidative stress possible
assay, TBARS assay (lipid 2016)
trol, rotenone only, rotenone and curcu- subcutaneously; 50 days) 2 injections intragastrically; via the Akt/Nrf2 signaling pathway.
peroxidation), spectrophot-
min-treated, and curcumin only groups) 50 days) Curcumin treatment regulated rota-
ometer (enzymatic activity)
tional behavior, and speed, time
and distance of movement were
improved.
PD Swiss albino male mice Rotenone Free Curcumin Greiss assay, Pyrogallol Curcumin increased GSH, SOD and (Khatri and
(n = 30; age: not specified; vehicle con- (1 mg/kg, 1 injection intraperitoneally; (pre- and co-treatment: 50, 100 and auto-oxidation assay, GSH CAT levels but decreased nitrite and Juvekar, 2016)
trol, rotenone only, rotenone and curcu- 21 days) 200 mg/kg orally; 1 h and 21 days) assay, spectrophotometry MDA levels; thereby inhibiting oxida-
min treated (50 mg/kg), rotenone and (MDA levels) tive stress. Curcumin also attenu-
curcumin treated (100 mg/kg), and rote- ated loss of locomotor function.
none and curcumin treated (200 mg/kg)
groups
PD Drosophila melanogaster male flies Paraquat Free Curcumin RT-PCR of oxidative Curcumin did not change the mRNA (Park et al.,

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


(n = 18; age: 2–3 days; control, para- (20 mM, soaked filter paper; 48 h) (pre-treatment: 0.02% solution on stress-related genes levels of SOD1, SOD2, CAT, 2012)
quat only, curcumin- and paraquat-trea- filter paper; 48 h pre-fed after GSTD1 and mth. Curcumin also alle-
ted, quercetin- and paraquat-treated 4 h starvation) viated locomotor dysfunction.
group, S. officinalis extracts and para-
quat-treated, and Z. rhizoma extracts
and paraquat-treated groups)
PD LRRK2-transgenic Drosophila melano- H2O2 Free Curcumin OxyBlot protein oxidation kit Curcumin reduced the levels of oxi- (Yang et al.,
gaster flies (0.1%; 2 weeks in feed), transgenic (co-treatment: 1 mM in feed; dized proteins and also improved 2012)
(gender: not specified; n = 30; age: not LRRK2 (UAS-LRRK2–1, 2 weeks) locomotor defects.
specified; DMSO control, H2O2 only, UAS-G2019S-2)
H2O2- and curcumin-treated, H2O2-
and LRRK2 inhibitor treated, curcumin
only, and LRRK2 inhibitor groups)
HD Wistar female rats 3-NP Encapsulated Curcumin, C-SLN TBA, DCHF-DA, dinitro- Curcumin nanoparticle, C-SLN, pro- (Sandhir et al.,
(n = 20; age: not specified; vehicle con- (20 mg/kg; 3 days) (post-treatment: 40 mg/kg daily; phenylhydrazone, fluoro- tected against oxidative stress due 2014)
trol, C-SLN-only, 3-NP only, and 3-NP 7 days) metric and hydroxylamine to the activation of the regulatory
and C-SLN-treated groups) autoxidation assays Nrf2 pathway. C-SLN also amelio-
rated neuromotor deficits.
SMA Transgenic AR97Q male mice Tg2576 APP transgenic Curcumin derivative, ASC-JM17 RT-PCR of Nrf1, Nrf2 and The curcumin derivative, ASC-JM17, (Bott et al.,
(n = not specified; age: 16 weeks; (overexpressing a mutant form (post-transfection:120 mg/kg; proteasome-encoding activated expression of both Nrf1 2016)
vehicle control, and transgenic and of human APP: K670 M/N671 L) 6 weeks) genes and Nrf2 which led to increased
ASC-JM17-treated groups) expression of downstream genes
coding for proteasome subunits.
ASC-JM17 also increased motor
function.
3-NP: 3-nitropropionic acid; 6-OHDA: 6-hydroxydopamine; Aβ40 or Aβ42: 40- or 42-amino acid amyloid-β peptide; Akt: serine/threonine protein kinase B; AlCl3: aluminum chloride; AD: Alzheimer's disease; APP: amyloid precursor protein; CAT:
catalase; DCHF-DA: dichlorodihydrofluorescein diacetate; DMSO: dimethyl sulfoxide; DNPH: 2,4-dinitrophenylhydrazine; DTNB: 5,5-dithio-bis-(2-nitrobenzoic acid) or Ellman's reagent; ELISA: enzyme-linked immunosorbent assay; GPx: glutathione
peroxidase; GR: glutathione reductase; GSH: glutathione; GSTD1: glutathione S-transferase D1; h: hours; H2O2: hydrogen peroxide; HD: Huntington's disease; HNE: 4-hydroxy-2-nonenal; HO-1: heme oxygenase 1; LC: liquid chromatography; LPS:
lipopolysaccharide; LRRK2: leucine-rich repeat kinase 2; MDA: malondialdehyde; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MS/MS: tandem mass spectrometry; mth: methuselah; Nrf1/2: nuclear factor-E2-related factor 1/2; PD: Parkinson's
disease; Plp1: proteolipid protein 1; ppm: parts per million; RT-PCR: real time polymerase chain reaction; SMA: spinal muscular atrophy; SOD: superoxide dismutase; SOD1/2: superoxide dismutase 1/2; TBA: thiobarbituric acid; TBARS: thiobarbituric
acid-reactive substances.

9
10
Table 4. Summary of the studies investigating the effect of curcumin in animal models of oxidative stress, dementia and aging.

Phenotype Animal model Toxin applied Curcumin treatment Assay/technique Main findings References
Oxidative Swiss Albino male mice Sodium nitroprusside Free Curcumin and encapsulated No oxidative assay used (only Theracumin protected against (Nazari et al.,
stress (n = not specified; age: 6 weeks; 9 (concentration: 10 nmol; dura- curcumin, Theracumin® indirect measure) motor dysfunction and neuronal 2014)
groups: vehicle control, and various tion: 24 h) (pre-treatment concentration: loss.

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


concentrations of curcumin and 100–200 mg/kg; duration: 24 h)
Theracumin®)
Oxidative Swiss Albino male mice LPS Free Curcumin TBARS and Griess assays Curcumin dose-dependently pre- (Jangra et al.,
stress (n = 48; age: not specified; 8 groups: (0.83 mg/kg; 3 h) (pre-treatment: 100, 200, 400 mg/kg, vented the increase in MDA and 2016)
untreated, and various concentra- orally ingested; 7 days) nitrates, and protected against
tions of LPS and curcumin) the reduction in GSH.
Oxidative Sprague–Dawley rats Aluminum Free Curcumin DTNB spectrophotometry Curcumin prevented depletion of (Sood et al.,
stress (gender: not specified; n = 40; age: (100 mg/kg; 8 weeks) (co-treatment: 50 mg/kg, GSH levels in the cerebral cortex, 2011)
not specified; control, aluminum intraperitoneal injection; 8 weeks) midbrain and cerebellum.
only, aluminum and curcumin, and
curcumin only groups)
Oxidative Wistar male rats Lead acetate Free Curcumin Thiobarbituric acid, serum TAC Curcumin inhibited lipid peroxida- (Hosseinzadeh
stress (n = 40; age: 8 weeks; control, lead (20 mg/kg; 8 weeks) (co-treatment: 30 mg/kg; 8 weeks) (hydroxyl radical) assays tion as indicated by reduced MDA et al., 2013)
acetate only, lead acetate and curcu- levels, while also increasing the
min, lead acetate and training, and total antioxidant capacity in plasma.
lead acetate, training and curcumin
groups)
Oxidative Wistar male rats Dopamine Free Curcumin Western blotting of AP-1 Curcumin protected against pro- (Luo et al.,
stress (n = not specified; age: 6 months; 6 (2 μmol; 24 h) (pre-treatment: 1–10 μmol; 24 h) longed activation of AP-1 and 1999)
groups: untreated and dopamine- thereby inhibited oxidative stress
treated groups at 3 different in the striatum.
concentrations)
Oxidative Swiss Albino male mice Kainic acid Free Curcumin Immunohistochemistry Curcumin reduced the number (Shin et al.,
stress (n = 28; age: 4 weeks; control, kainic (30 mg/kg; 48 h) (pre-treatment: 200 mg/kg; 24 h) of HO-1 positive cells in the 2007)
acid, curcumin and kainic acid, and hippocampus.
curcumin only groups)
Oxidative Wistar male rats Kainic acid Free Curcumin and curcuminoids, Western blotting of nitrosative Curcuminoids prevented the (Sumanont
stress (n = 56; age: 9–10 weeks; vehicle (10 mg/kg) diacetylcurcumin and manganese stress-related protein, increase of nitric oxide levels and et al., 2007)
control, curcumin and kainic acid (pre-treatment: 50 mg/kg; 24 h) immunohistochemistry mRNA expression of the iNOS
treated, and curcuminoids and kainic gene.
acid treated groups)
Oxidative Wistar female rats Parathion Free Curcumin Colorimetric assay Curcumin suppressed MDA (Canales-Aguirre
stress (n = 40; age: not specified; control, (inhalation; 2 months) (pre-treatment: 200 mg/kg; levels in hippocampus. et al., 2012)
parathion only, curcumin and para- 2 months)
thion, and curcumin only groups)
Phenotype Animal model Toxin applied Curcumin treatment Assay/technique Main findings
Oxidative Swiss Albino male mice Fluoride Free Curcumin Trichloroacetic acid (TCA) assay:
Curcumin protected against the (Sharma et al.,
stress (n = 28; age: 1.5 months; control (120 ppm; 30 days) (co-treatment: 30 mg/kg b.w.; 15% w/v thiobarbituric acid increase in MDA in the 2014)
and 3 experimental groups) 30 days) (TBA), 0.375% hydrochloric acid hippocampus.
Oxidative Male rats Nicotine Free Curcumin Thiobarbituric acid assay, absor-
Curcumin dose-dependently (Motaghinejad
stress (type: not specified; n = 60; age: (6 mg/kg/day, subcutaneous (co-treatment: 10, 20, 40 and 60 mg/ bance, NADPH assay decreased MDA, increased et al., 2017)
8 weeks; untreated, nicotine treated, injection; 21 days) kg, intraperitoneal injection; 21 days) GSH, decreased GSSG, and
and nicotine and curcumin treated increased SOD, GPx and GR
groups) enzyme activities. Curcumin
also prevented behavioral
defects.
Oxidative White New Zealand male rabbits High fat dietA Free Curcumin Griess assay, LDH kit, spectro- Curcumin increased SOD activity (Manogaran
stress (n = 16; age: not specified; control, (co-treatment: 80 mg/kg; 16 weeks) photometry (NBT) and prevented lipid peroxidation et al., 2015)
high fat diet, simvastatin treated
and high fat diet, and curcumin trea-

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


ted and high fat diet groups)
Dementia Wistar male rats Streptozotocin Free Curcumin Colorimetric assay, spectrophoto- Curcumin inhibited lipid peroxida- (Ishrat et al.,
(n = 40; age: 1 year; sham-oper- (1.5 mg/kg b.w.; incubation: (post-treatment: 80 mg/kg; 3 weeks) metry (trichloroacetic acid) tion and stimulated GSH-linked 2009)
ated, vehicle-treated control, sham- single dose) antioxidant activity, and led to
operated and curcumin-treated, the inhibition of STZ-induced cog-
ICV-STZ-infused and vehicle-treated nitive decline in the hippocampus
lesioned, and ICV-STZ-infused and and cerebral cortex.
curcumin- treated groups)
Dementia Wistar male rats Streptozotocin Free Curcumin Colorimetric assay Curcumin suppressed the increase (Samy et al.,
(n = 32; age: adult; untreated, ery- (3 mg/kg; single dose) (post-treatment: 80 mg/kg/day; in MDA and depletion of GSH, 2016)
thropoietin treated, curcumin treated, 3 months) thereby inhibiting lipid peroxidation
and both erythropoietin and curcu- and promoting antioxidant activity.
min-treated groups)
Dementia Sprague Dawley male rats Streptozotocin Free Curcumin ELISA assay (tetra-ethoxy pro- Curcumin prevented the increase (Agrawal et al.,
(n = 40; age: adult; 8 groups: (3 mg/kg; 14 days) (pre- and post-treatment: pane for MDA; 2-nitro-benzoic in MDA and the decrease in GSH 2010)
untreated and various combinations 200 mg/kg; 1 h) acid for GSH) in the hippocampus and cortex.
of curcumin and STZ groups)
aging Laca male mice D-galactose Free Curcumin Thiobarbituric acid, Griess assays, Curcumin prevented the increase (Kumar et al.,
(n = 72; age: 2–3 months; vehicle, (100 mg/kg; 6 weeks) (co-treatment: 15 and 30 mg/kg, DTNB spectrophotometry in MDA and nitrites. Curcumin also 2011)
D-galactose, low dose curcumin and orally ingested; 6 weeks) protected against the reduction of
D-galactose, high dose curcumin and GSH levels, as well as SOD, GST
D-galactose, and curcumin groups) and CAT enzyme activities.
aging Wistar rats D-galactose Free Curcumin DNPH spectrophotometry, FRAP Curcumin increased the total anti- (Banji et al.,
(gender: not specified; n = 42; age: (150 mg/kg; 56 days) (pre- and co-treatment: 20 and assay oxidant capacity and inhibited 2013)
16 weeks and 20 months; 7 groups: 40 mg/kg; 60 days) and piperine protein oxidation. Curcumin also
vehicle controls, various concentra- (pre- and co-treatment: 7.5 and protected against motor
tions and combinations of curcumin 15 mg/kg; 60 days) dysfunction.
and piperine)
(continued on next page)

11
12
Table 4 (continued)
Phenotype Animal model Toxin applied Curcumin treatment Assay/technique Main findings References
aging Wistar rats D-galactose Free Curcumin DTNB spectrophotometry, Curcumin inhibited MDA, carbo- (Banji et al.,
(gender: not specified; n = 42; age: (150 mg/kg; 56 days) (pre- and co-treatment: 50 and thiobarbituric acid assay nyls and advanced oxidation pro- 2014)
16 weeks; 6 groups: vehicle con- 100 mg/kg; 63 days) and hesperidin tein products in the hippocampus.

Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21


trols, various concentrations and (pre- and co-treatment: 10 and
combinations of curcumin and 25 mg/kg; 63 days)
hesperidin)
aging Kunming mice D-galactose Free Curcumin TBA assay Curcumin prevented the decrease (Fu et al., 2017)
(gender: not specified; n = 4; age: (conditions not specified) (pre-treatment: 10 and 150 mg/kg; in GPx, SOD and CAT enzyme
not specified; vehicle control group, 7 weeks) activities, and increase in MDA
D-galactose only group, curcumin levels when compared to the
only, and D-galactose and curcumin D-galactose group.
groups)
aging SAMP8 and SAMR1 male mice SAMP8 and SAMR1 strains Free Curcumin TBARS assay, SOD commercial Curcumin reduced MDA levels (Sun et al.,
(n = 88; age: 6 months; SAMR1 (senescence-accelerated-resis- (post-transfection: 20 and kits and increased SOD activity in 2013)
untreated, SAMP8 untreated, and tant strain mice) 50 mg/kg b.w.; 25 days) the hippocampus.
SAMP8 mice and curcumin treated
groups)
aging Wistar Albino female rats None Free Curcumin Western blotting of nitrosative Curcuminoids down-regulated (Rastogi et al.,
(n = 24; age: 2–3 months and (only treatment: 100 mg/kg; stress-related proteins neuronal NOS in mitochondria. 2014)
24–25 months; young control, aging 3 months)
control, curcuminoid-treated young,
and curcuminoid-treated aging
groups)
AP-1: activated protein 1; b.w.: body weight; CAT: catalase; DTNB: 5,5-dithio-bis-(2-nitrobenzoic acid) or Ellman's reagent; ELISA: enzyme-linked immunosorbent assay; FRAP: ferric reducing antioxidant power; GSH: glutathione; GST:
glutathione S-transferase; h: hours; ICV-STZ: intracerebroventricular–streptozotocin; iNOS: inducible nitrogen oxide synthase; HO-1: heme oxygenase 1; LPS: lipopolysaccharide; MDA: malondialdehyde; ppm: parts per million; SAMP8:
senescence accelerated mouse-prone 8; SAMR1: senescence-resistant 1 mice; SOD: superoxide dismutase; TAC: total antioxidant capacity; TBA: thiobarbituric acid; TBARS: thiobarbituric acid-reactive substances; TCA: trichloroacetic
acid assay; w/v: percent weight/volume; w/w: percent mass.
A
bengal gram (30% w/w), sucrose (25% w/w), whole milk powder (16% w/w), yeast (1% w/w), hydrogenated groundnut oil (10% w/w), cholesterol (5% w/w), sodium chloride (5% w/w), shark liver oil (2% w/v) and egg yolk powder (7% w/w).
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 13

Eleven studies used toxin-treated or transfected cellular mod- (Mythri et al., 2011), therefore providing indirect support for
els of PD to investigate the therapeutic effect of curcumin the anti-nitrosative stress effect of curcumin.
against oxidative stress (Table 1). In 2 studies, SH-SY5Y Notably, in contrast to the other studies on cellular models
and SK-N-SH neuroblastoma cells were pre-treated with curcu- of PD, Ortiz-Ortiz et al. (2009) showed that curcumin
min derivatives and thereafter exposed to rotenone, an estab- increased ROS production in paraquat-exposed N27 cells.
lished model for PD (Jayaraj and Tamilselvam, 2013; Although curcumin did not reduce cell viability, curcumin
Ramkumar et al., 2017). In both studies, the authors con- pre-treatment increased levels of markers of apoptosis.
cluded that rotenone-induced oxidative stress was sup- However, the vehicle control for curcumin was not tested in
pressed by the curcumin derivatives, and thereby apoptosis this study, possibly explaining these conflicting results.
was reduced. Another study pre-treated SH-SY5Y cells with Ortiz-Ortiz et al. (2009) also showed that curcumin protected
curcumin followed by paraquat exposure to mimic neuronal against the degradation of Iĸβ, prevented the translocation
damage observed in NDDs (Jaroonwitchawan et al., 2017). of NFĸ-β, and thereby suppressed nitrosative stress in
Curcumin reduced ROS levels in paraquat-exposed cells paraquat-treated N27 cells. The authors implied that curcumin
and increased expression of the antioxidant genes, SOD may act via the NFĸ-β signaling pathway to protect against
and GPx. In two additional studies, SH-SY5Y and PC12 cells nitrosative stress.
were transfected with mutant α-synuclein and cells were incu-
bated post-transfection with curcumin (Wang et al., 2010; Liu Spinal muscular atrophy
et al., 2011). Both studies found that curcumin attenuated SMA is an autosomal recessive disorder, related to NDDs,
α-synuclein-mediated ROS production. However, it should with mutations or deletions in the survival of motor neuron
be noted that curcumin had no effect on ROS when the cells (SMN) gene. Lack of the SMN protein causes degeneration
were not induced with mutant α-synuclein, which might indi- and results in clinical evidence of anterior horn cell dysfunc-
cate that the protective properties of curcumin may only occur tion (Lunn and Wang, 2008). Although SMA pathogenesis
in response to an oxidative stressor. has been linked to dysfunctional SMN protein processing,
One study exposed N27 neuronal cells to buthionine oxidative damage has also been proposed as a possible
sulfoximine to inhibit GSH, proposed as an early trigger of mechanism underlying SMA pathology.
oxidative stress in PD (Harish et al., 2010). Cells were treated Currently, there is only 1 study in which the antioxidant
with curcumin and 3 bioconjugates before and after exposure properties of curcumin were studied in a cellular model of
to buthionine sulfoximine. Curcumin and its bioconjugates were SMA (Table 1). Köstel et al. (2012) showed that curcumin
shown to reduce levels of ROS, MDA and H2O2 while only protected SMA patient-derived fibroblasts against oxidative
curcumin, additionally, increased GSH and glutathione stress by reducing ROS levels. Interestingly, curcumin
S-transferase (GST) protein levels. It should also be noted increased ROS in fibroblasts from both healthy controls and
from the study's findings that the structural alterations of the cur- SMA carriers. The authors proposed that the basal oxidative
cumin bioconjugates affected its protective ability. stress levels are lower in the controls and SMA carriers, com-
Four studies used MPP + (1-methyl-4-phenylpyridinium) pared to SMA patients, and therefore curcumin was believed
treatment as a cellular model of PD. In one, primary rat astro- to act as a pro-oxidant in the controls and carriers. However,
cytes were pre-treated with curcumin and subsequently ex- the antioxidants vitamin E and N-acetylcysteine, which were
posed to the toxin (Yu et al., 2016). Curcumin reduced the also investigated in this study, decreased ROS in both
protein levels of ROS, TNF-α and IL-6 while restoring GSH controls and carriers regardless of the basal ROS levels.
levels and thereby inhibited oxidative stress. Curcumin also
reduced TLR4, MyD88 and TRIF protein levels, therefore impli- Multiple sclerosis
cating the TLR4 immune response pathway in the curcumin- MS is a disorder in which there are features of progressive
mediated antioxidant response. In a further study, in which neurological dysfunction, in which the immune system
PC12 cells were co-treated with both curcumin and MPP +, it damages the myelin sheath in the brain and spinal cord
was found that curcumin reduced MPP +-mediated ROS (Benn et al., 2001). MS affects several cells with myelin-
levels, resulting in the prevention of apoptosis and mitochon- producing oligodendrocytes being the major affected cell
drial membrane instability (Chen et al., 2006). Curcumin was type. Oligodendrocytes are vulnerable to oxidative stress
also shown to reduce iNOS levels and thereby prevent the and produce RNS, which in turn affects demyelination of
toxic overexpression of RNS. Pre-treatment of curcumin neurons.
improved cell viability in the MPP +- and H2O2-mediated Only 1 study has investigated the antioxidant effects of curcu-
PC12 cell models of PD (Marchiani et al., 2013). Although min in MS (Table 1). The authors investigated the effects of
free radical levels were not assessed in PC12 cells, the several dietary compounds, including curcumin, on H2O2-
cell-free experiments in this study showed that curcumin exposed oligodendrocytes (OLN-93) and lipopolysaccharide
and its derivatives had a high affinity for scavenging free radi- (LPS)-exposed macrophages (Van Meeteren et al., 2004).
cals. In another study, N27 cells underwent a curcumin pre- Notably, curcumin did not protect against H2O2-induced
treatment followed by MPP + exposure (Mythri et al., 2011). oxidative damage and LPS-induced nitrosative stress in
Although curcumin improved cell viability, no nitrosative these cells. The authors speculated that the poor efficacy of
stress markers were investigated. Additionally, cell-free curcumin observed in their study, compared to other studies,
experiments of isolated mitochondria showed that curcumin could be due to the differences in cell lines, macrophage
and a curcumin derivative reduced peroxynitrate levels stimulation and the source of curcumin used.
14 Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

Antioxidant effects of curcumin in models of Lipopolysaccharide-induced model


oxidative/nitrosative stress In LPS-induced oxidative stress, mouse N9 microglial cells
were pre-treated with curcumin or a curcumin derivative and
We identified 14 studies which investigated the effect of
subsequently exposed to LPS (Zhang et al., 2010). The
curcumin in cellular models of oxidative stress, which
derivative dose-dependently decreased levels of ROS, nitrite
were linked to neurodegeneration in general and not a
(NO −) and iNOS in cells. Moreover, curcumin and its deriva-
specific type of NDD (Table 2). No studies, to date, have
tive inhibited the LPS-induced phosphorylation of IκBα (NF-κβ
been performed on models of nitrosative stress. More details
pathway), ERK1/2, p38 MAPK, and JNK (MAPK pathway).
on each study are provided below but in summary, all of the
studies showed significant antioxidant effects of curcumin. Metal ion-induced model
Curcumin treatment was mainly administered prior to the Rat cortical neurons were pre-treated with curcumin followed
toxin exposure (pre-treatment), with a wide range of toxins by exposure to copper, Cu(II), to induce oxidative damage
and various human and animal cell lines (both primary and (Huang et al., 2011). Curcumin dose-dependently reduced
immortalized) were used. Again, this indicates that curcu- ROS and H2O2 levels, thereby inhibiting oxidative stress.
min's therapeutic properties are observed at various concen- The authors proposed that the antioxidant effects of curcumin
trations of different toxins and acting on a wide range of prevented Cu(II)-induced apoptosis. In curcumin pre-treated
cell types. Neuro-2A and HEK293 cells exposed to iron, Fe 2+ (Morales
et al., 2017), curcumin protected against oxidative damage
Hydrogen peroxide-induced model induced by iron exposure.
To date, 5 studies have used H2O2 exposure as a model. In
Iodoacetate-induced model
1 study, glioblastoma A172 and primary spinal cord HA-sp
Iodoacetate inhibits the glycolytic enzyme glyceraldehyde 3-
astrocytic cells were both pre- and post-treated with curcumin
phosphate dehydrogenase (GAPDH) that leads to oxidative
(Daverey and Agrawal, 2016), with the finding that curcumin
stress (Schmidt and Dringen, 2009). Rat cerebellar granule
treatment protected cells against oxidative stress, including
neurons were pre-treated with curcumin followed by expo-
the disruption of cytonemes and F-actin aggregates.
sure to iodoacetate (Reyes-Fermin et al., 2012). Curcumin
The neurodegenerative astrocyte activation was inhibited
reduced iodoacetate-induced ROS levels in cells. Indepen-
in curcumin-treated cells. Moreover, curcumin prevented
dently of iodoacetate toxicity, curcumin dose-dependently
alterations in mitochondrial morphology following oxidative
increased heme oxygenase 1 activity in healthy cells.
stress. Another study pre-treated PC12 cells with curcumin,
which was then followed by H2O2 exposure (Fu et al., Hypoxia-induced model
2016), with results demonstrating that curcumin improved cell Chhunchha et al. (2013) pre-treated HT22 mouse hippocam-
viability and prevented DNA damage and apoptosis. Further- pal cells with curcumin followed by hypoxia-induced oxidative
more, the authors showed that the protective action of curcu- stress. Curcumin pre-treatment reduced ROS expression
min acted via the cell growth-death MAPK (mitogen-activated when compared to hypoxia exposure only. Curcumin pre-
protein kinases) and Akt (serine/threonine protein kinase) vented downregulation of the cytoprotective peroxiredoxin
pathways. Jiang et al. (2011) pre-treated astrocytes with cur- (Prdx6) protein which led to reduced ROS levels, and thereby
cumin before H2O2 exposure. The astrocytes were obtained attenuated hypoxia-induced damage.
from mice with (Nrf2 +/+) and without the antioxidant-inducer
Nrf2 (Nrf2 −/−). Curcumin reduced ROS production in Nrf2 +/+ Excitotoxicity model
astrocytes but not in Nrf2 −/−. Additionally, curcumin increased Glutamate upregulation has been linked to excitotoxicity and
expression of the antioxidant genes and proteins, heme oxidative stress (Albrecht et al., 2010). Exposure to gluta-
oxygenase 1 (HO-1), glutamate cysteine ligase (GCL) and mate was used in 2 studies using a PC12 cell model. In 1
quinine oxidoreductase 1 (NQO1), but only in Nrf2 +/+ cells. study, the neuroprotective effects of curcumin were tested
However, it should be noted that only the findings between in a glutamate hyperstimulated PC12 cell model (Chang
curcumin-treated and untreated groups were statistically sig- et al., 2014). Curcumin reduced GSH, SOD and nitric oxide
nificant which constrain the interpretation of the results. In a protein levels, which led to reduced cell death. It is unclear
Neuro-2A cell model, cells were pre-treated with curcumin whether curcumin-mediated reduction in SOD levels pro-
followed by H2O2 exposure (Zhao et al., 2011). Curcumin moted antioxidant activity. However, the reduction in nitric
reduced ROS and calcium, and inhibited the IκBα degrada- oxide indicates that curcumin reduced nitrosative stress in
tion and NF-κB nuclear translocation from the cytosol. Finally, PC12 cells. Overall, the authors claimed that curcumin's pro-
1 study (Öz and Çelik, 2016) transfected SH-SY5Y cells with tective effects acted via GSH and mitochondria-dependent
transient receptor potential melastatin-like 2 (TRPM2) chan- nitric oxide–ROS pathways. In the second study, cells were
nel protein, and subsequently treated with curcumin and pre-treated with the curcumin derivative, J147, and thereafter
H2O2. Curcumin inhibited the H2O2-induced activation of exposed to either glutamate or tert-butyl hydroperoxide
TRPM2, and reduced TRPM2 protein levels. Curcumin also (t-BHP) (Chen et al., 2015). J147 reduced the levels of ROS
decreased ROS levels in transfected cells (vs. untreated and RNS specifically H2O2, OH, nitric oxide and peroxynitrite.
transfected) and non-transfected cells (vs. untreated non- One study used N-methyl- -aspartic acid (NMDA) as a model
transfected). The authors concluded that curcumin reduced of oxidative-inducing excitotoxicity, in which rat neuronal and
apoptosis and inhibited oxidation via the TRPM2 channels. astrocytic cells were treated with curcumin and thereafter
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 15

with NMDA (Lin et al., 2011). Although curcumin inhibited prior to LPS injection, which elicited inflammation and oxida-
iNOS expression under non-stressed conditions, iNOS tive damage. Both models of curcumin administration had
expression was not measured under NMDA-toxic conditions similar effects. Curcumin and its derivative prevented the
and therefore the anti-nitrosative effects of curcumin were increase of isoprostanes and iNOS mRNA. However, only
not analyzed. curcumin treatment suppressed the protein carbonyl and
nitrotyrosine levels. Seo et al. (2010) fed transgenic Tg2576
‘Non-toxin’ model amyloid mice with curcumin for 6 months and demonstrated
As opposed to studies using induction of oxidative stress, suppression of lipid peroxidation in the spinal cord. It was
Panchal et al. (2008) analyzed gene expression in rat glioma suggested that the curcumin-mediated reduction of lipid per-
C6 cells following curcumin treatment under non-externally oxidation in the spinal cord led to the rescue against demye-
stressed conditions. Eight main clusters were identified from lination and neuronal loss in mice. J147 curcumin derivative
the Hierarchical Ordered Partitioning and Collapsing Hybrid was administered, for 5 months, to amyloid transgenic mice
clustering (HOPACH) analysis of 1223 genes differentially (Chen et al., 2011), and it was shown to prevent an increase
expressed between curcumin-treated and control cells. The in heme oxygenase 1 levels. The authors stated that J147
oxidative stress pathway was one of the pathways with a high uniquely acted on several disease mechanisms involved in
number of differentially expressed genes. Curcumin treat- AD, namely oxidative damage, inflammatory response,
ment was shown to increase the mRNA expression of the amyloid-β deposition and neuronal loss.
glutathione S-transferase A5 (GSTA5) and heme-oxygenase In a rat model of AD, rats were pre-treated with curcumin
1 (HMOX1) genes, which are both involved in oxidative for 2 months prior to intraventricular injection of amyloid-β
stress. The quinine oxidoreductase 1 (NQO1), catalase (Frautschy et al., 2001). Curcumin protected against the
(CAT), microsomal glutathione S-transferase 1 (MGST1), increase in isoprostane levels in the cortex, and therefore
and superoxide dismutase 2 (SOD2) genes involved in the suppressed fatty acid oxidation. In another study, rats were
antioxidant response were also increased in curcumin- injected with aluminum chloride (AlCl3) to induce AD symp-
treated cells. toms and were orally post-treated with a novel curcumin
derivative, CUR-CA-THIONE (Shelat and Acharya, 2016).
STUDIES PERFORMED IN ANIMAL MODELS This compound inhibited AlCl3-induced increase in lipid
peroxidation. Furthermore, although it increased GSH and
Antioxidant effects of curcumin in catalase protein levels it also reduced SOD levels. The
neurodegenerative disorders authors provide no explanation for the conflicting finding in
In addition to cellular models, 14 studies investigated the anti- which CUR-CA-THIONE increased protein levels of some
oxidant properties of curcumin in toxin-treated or transgenic antioxidant enzymes (GSH and catalase) but reduced levels
animal models of NDDs (Table 3). There were 6 studies of other antioxidants (SOD). The duration of exposure and
performed on AD, 6 on PD and 1 each on HD and SMA. On the intensity of the oxidative stressors are factors which
review of these studies, there appears to be an overall consen- influence whether a decrease or increase in the antioxidant
sus that curcumin, or derivatives thereof, suppressed toxin- enzyme levels is observed, and this might explain the discre-
induced oxidative stress in animal models. In addition to anti- pancies observed.
oxidant effects, curcumin treatment improved balance, speed
of movement and coordination. The animal models included Parkinson's disease
transgenic and various toxin-induced models, as well as a Six studies investigated the antioxidant effect of curcumin in
range of animals including rats, mice and Drosophila. How- animal models of PD (Table 3). Of these, 2 used rotenone
ever, some of these studies were constrained by the lack of and the rest used 6-OHDA, MPTP, paraquat or H2O2 (1 study
cytotoxicity analysis of curcumin, absence of properly con- for each) to elicit a PD phenotype. Curcumin and its deriva-
trolled groups and small sample sizes which may account tives were pre-treated orally prior to the neurotoxic 6-OHDA
for some of the contradictory findings between the studies. striatal injection in rats (Agrawal et al., 2012). Curcumin
and its derivatives prevented the 6-OHDA-induced reduction
Alzheimer's disease in the GSH, GPx, GR, and SOD and catalase enzyme
Six studies reported curcumin's effect in animal models of AD levels. In addition, curcumin and its derivatives reduced lipid
(Table 3). Four of these used transgenic animals over- peroxidation as measured by reduced MDA levels. There-
expressing amyloid precursor protein. In 1 study, amyloid pre- fore, curcumin reduced oxidative stress, a finding associated
cursor protein Tg2576 transgenic mice were fed with low- and with improved motor function and inhibition of neuronal loss
high-dose curcumin-containing chow for 6 months (Lim et al., in the 6-OHDA treated rats. Notably, curcumin or its deriva-
2001). Both curcumin dosages suppressed levels of oxidized tives did not restore the antioxidant activity to the untreated
protein carbonyls up to 60%. The authors suggested that level. It would, therefore, be interesting to determine the
curcumin is effective at low dosages and that the inhibition duration of protection afforded by curcumin.
of protein oxidation may be responsible for the suppression In a study by Jayaraj et al. (2014), C57BL/6 mice were pre-
of amyloid-β accumulation in mice. In another mouse model, treated with the curcumin derivative (CNB-001) prior to MPTP
Tg2576 amyloid transgenic mice were fed curcumin and its exposure followed by co-treatment for 4 to 7 days thereafter.
derivative in chow for 4 months (Begum et al., 2008). A sepa- This study showed that the MPTP only group developed
rate group of mice received oral gavage of curcumin 2 days locomotive and exploratory behavior deficits and impaired
16 Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

GSH levels. The CNB-001- and MPTP-treated group was in solid lipid nanoparticles. C-SLN prevented the increase in
noted to have inhibition of oxidative stress as evidenced by MDA, ROS and protein carbonyls. Moreover, it protected
the reduced lipid peroxidation and increased levels of GSH against the depletion of GSH, while increasing SOD activity.
and GPx. Contrary to other studies; the combination of Rats treated with C-SLN also showed an increase in the
CNB-001- and MPTP-treatments suppressed the levels of expression of the antioxidant-inducing Nrf2.
protective SOD and catalase enzymes. It was proposed that
the increased SOD and catalase levels were due to an adap- Spinal muscular atrophy
tive response to the oxidation-inducing MPTP. The antioxidant effect of curcumin was investigated in 1 study
In a rotenone-mediated PD model, rats were initially treated of an animal model (mouse) of SMA (Table 3). In the trans-
with curcumin for 4 days and thereafter co-treated with curcu- genic AR97Q mouse model, the ASC-JM17 curcumin analo-
min and rotenone for 50 days (Cui et al., 2016). Similarly to gue was given via oral gavage for 6 weeks (Bott et al., 2016).
the 6-OHDA model, curcumin treatment suppressed the ASC-JM17 activated expression of both Nrf1 and Nrf2 which
rotenone-mediated oxidative stress by elevating GSH levels, led to increased expression of downstream genes coding for
while reducing ROS and MDA levels. The study also showed proteasome subunits, molecular chaperones and antioxidant
that curcumin's antioxidant effects seemed to act via the enzymes; and thereby improved motor function in the trans-
Akt/Nrf2 signaling pathway, due to the phosphorylation of genic mice.
Akt and Nrf2, and the upregulation of heme oxygenase 1 and
quinine oxidoreductase 1 protein expression. Another study
Antioxidant effects of curcumin in models of
investigated curcumin's effect in the rotenone-mediated PD
oxidative/nitrosative stress
model with a shorter treatment duration of 21 days in Swiss
albino mice (Khatri and Juvekar, 2016). Curcumin dose- Eleven studies tested the antioxidant effects of curcumin in
dependently prevented the rotenone-induced increase in models of oxidative stress in animals treated with toxins,
MDA and nitrite levels, while preventing the decrease in metal ions and excitotoxic inducers (Table 4). No studies
GSH, SOD and catalase levels. However, it should be noted were performed specifically on nitrosative stress. All of these
that there was no curcumin only treatment group, a limitation studies found that curcumin or curcumin derivatives showed
which constrains interpretation of the results. antioxidant protection in various animals including mice, rats
Two studies used Drosophila melanogaster as a model. In and rabbits. However, various assays or techniques were
1 study, the flies were pre-fed curcumin followed by paraquat used making it difficult to compare findings between studies.
exposure (Park et al., 2012). Paraquat caused locomotor
dysfunction, increased mortality and reduced expression of Sodium nitroprusside-induced model
antioxidant genes. Although curcumin improved survival Sodium nitroprusside is a vasodilator which stimulates oxida-
and locomotor deficits, it did not upregulate the expression tive stress (Nazari et al., 2012), and in a mouse model of
of antioxidant genes. This may indicate that in flies curcumin sodium nitroprusside-induced oxidative stress, animals were
may act via another pathway, and not the antioxidant orally pre-treated with curcumin and the curcumin derivative,
pathway. In a transgenic Drosophila LRRK2 model of PD, Theracumin (Nazari et al., 2014). Theracumin protected
flies were co-fed curcumin and H2O2 (Yang et al., 2012). against motor dysfunction and neuronal loss. No oxidative
Curcumin reduced the total oxidized protein levels in both markers or antioxidant agents were tested and therefore the
LRRK2 wild type and LRRK2 mutant flies fed H2O2. It was antioxidant effects of curcumin and Theracumin were inferred
found that curcumin improved survival rates of the flies and from the prevention of sodium nitroprusside-induced apoptosis.
inhibited mutant LRRK2 activity. This study showed that
curcumin protected against oxidative stress but also may be Lipopolysaccharide-induced model
an LRRK2 kinase inhibitor. In an LPS-induced mouse model, mice were orally treated
with curcumin for 7 days prior to LPS injection (Jangra et al.,
Huntington's disease 2016). Curcumin decreased levels of MDA and nitrates, and
HD is a monogenic, autosomal dominant disorder in which increased GSH levels. All protective effects of curcumin had
pathogenic CAG repeat expansions in the HTT gene result in a larger effect size when curcumin was co-administered
the production of mutant huntingtin protein (The with piperine, to increase its bioavailability (Suresh and
Huntington's Disease Collaborative Research, 1993). The Srinivasan, 2010).
symptoms of HD include progressive neuropsychiatric distur-
bance and chorea (Montoya et al., 2006). Similarly to PD and Metal ion-induced model
AD, HD has also been associated with mitochondrial dys- In an aluminum-induced rat model of oxidative stress, rats
function and oxidative stress as possible disease mechan- were intraperitoneally co-treated with curcumin and aluminum
isms (Arun et al., 2016). (Sood et al., 2011). Curcumin prevented the depletion of GSH
To date, only 1 study has analyzed the antioxidant proper- levels and increased mRNA expression of the molecular
ties of curcumin in an animal (rat) model of HD (Table 3). In chaperone heat shock protein, HSP70. It was suggested that
this model, rats were injected with the ROS-producing neuro- the improvement in mitochondrial activity was due to the
toxin, 3-nitropropionic acid (Sandhir et al., 2014). The rats antioxidant effect of curcumin. In a lead-induced rat model,
were subsequently treated by oral gavage with curcumin rats were co-treated with curcumin and lead intraperitoneally
nanoparticles (C-SLN), in which curcumin was encapsulated for 8 weeks (Hosseinzadeh et al., 2013). Curcumin inhibited
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 17

lipid peroxidation, while also increasing the total antioxidant To date, only studies on animal models on dementia and
capacity. aging have been performed, and these have been included
in Table 4. To summarize, all 9 studies showed an anti-
Excitotoxicity model oxidant effect of curcumin against lipid and protein oxidation
Six studies were performed using different neurotoxins in animals, including mice and rats. For these studies, only
including dopamine, kainic acid, parathion, fluoride and streptozotocin was used to induce dementia, whereas D-
nicotine. In the dopamine-induced rat model of excitotoxicity galactose, transgenic mice and natural aging were used to
and oxidative stress, rats were pre-treated with curcumin induce features of aging.
followed by dopamine exposure (Luo et al., 1999). Curcumin To date, only 3 studies have investigated the antioxidant
protected against prolonged activation of activated protein properties of curcumin in a streptozotocin-induced rat model
1 (AP-1) and therefore, it was proposed that oxidative stress of dementia (Table 4). Two of the studies only post-treated
and apoptosis were suppressed. Kainic acid is an oxidative rats with curcumin (Ishrat et al., 2009; Samy et al., 2016),
agent which stimulates oxidative stress and leads to neuro- while the third study pre- and post-treated rats (Agrawal
toxicity. Two studies investigated a kainic acid-induced rat et al., 2010). Both pre- and post-treatments of curcumin
model in which animals were intraperitoneally injected with suppressed the increase in MDA and depletion of GSH in
various curcuminoids and thereafter exposed to kainic acid all 3 studies. Furthermore, Ishrat et al. (2009) showed that
(Shin et al., 2007; Sumanont et al., 2007). Curcuminoids curcumin prevented the increase in 4-hydroxy-2-nonenal,
reduced nitric oxide levels and expression of the iNOS gene. TBARS, protein carbonyls and H2O2, while increasing the
A mouse model was treated with an intraperitoneal injection activity of antioxidant enzymes GPx and GR.
of curcumin prior to kainic acid injection (Shin et al., 2007). Six studies have investigated animal models of aging and
Curcumin reduced the number of heme oxygenase 1 positive the antioxidant effects of curcumin (Table 4). Four of the 6
hippocampal cells. The authors proposed that the antioxidant studies used mouse and rat models of aging in which animals
effects of curcumin played a role in the suppression of apop- were treated orally or by injection with curcumin and aging-
tosis. In a parathion model of neurotoxic-mediated oxidative inducing D-galactose (Kumar et al., 2011; Banji et al., 2013,
stress, rats were treated with curcumin for 1 week prior to 2014; Fu et al., 2017). In these D-galactose studies, curcumin
parathion exposure (Canales-Aguirre et al., 2012). Curcumin prevented an increase in MDA, nitrites, and protein carbonyls
inhibited the increase in MDA in the hippocampus. It should and thiols. Curcumin also protected against the reduction of
be noted that curcumin's antioxidant ability may play a role GSH levels, and SOD, GST, GPx and catalase enzyme
in curcumin preventing apoptosis and microgliosis in activities. In a transgenic SAMP8 mouse model of accelerated
parathion-induced rats. In a fluoride-exposed mouse model aging, curcumin was administered intragastrically for 25 days
of neurotoxic-mediated oxidative stress, curcumin and (Sun et al., 2013). Curcumin reduced MDA levels and
fluoride were orally co-treated for 30 days (Sharma et al., increased SOD activity in mice hippocampus. Nineteen-
2014). Curcumin protected against an increase in MDA in month-old rats were used as a model for aging with the rats
the hippocampus. Finally, in a nicotine-induced rat model, fed curcuminoids for 3 months (Rastogi et al., 2014). The
rats were intraperitoneally co-treated with curcumin and nico- curcuminoids lowered neuronal NOS levels in the aged rats.
tine (Motaghinejad et al., 2017). Curcumin dose-dependently The authors concluded that the improvement in aging-
prevented an increase in MDA, depletion of GSH, increase in induced mitochondrial impairments was partly influenced by
GSSG, and decrease in SOD, GPx and GR enzyme activities. curcumin inhibition of neuronal NOS.

Neuroinflammatory-induced model
In a cholesterol-induced rabbit model of neuroinflammation to CURCUMIN-MEDIATED ANTIOXIDANT PATHWAYS
elicit oxidative damage, rabbits were orally co-treated with DERIVED FROM THE REVIEWED STUDIES
curcumin and a high fat diet (Manogaran et al., 2015). Curcu- The studies which have been reviewed have shown that cur-
min increased SOD activity and prevented lipid peroxidation. cumin acted via multiple pathways to block oxido-nitrosative
Curcumin also reduced plasma and brain cholesterol levels, stress (Tables 1–4). Fig. 1 summarizes the information gath-
possibly due to increased antioxidant activity. ered from the reviewed articles and depicts the main findings
from these studies. Curcumin was shown mainly to inhibit
lipid and protein oxidation with a reduction in levels of MDA,
ANTIOXIDANT EFFECTS OF CURCUMIN IN as well as protein carbonyls, thiols and nitrotyrosines. Curcu-
MODELS OF DEMENTIA AND AGING
min also inhibited unregulated DNA damage by stimulating
Additionally, we included studies involving curcumin in Akt and MAPK pathways (Fig. 1). The activities of the anti-
dementia and aging since these have both been linked to oxidant enzymes, specifically SOD, catalase, GPx, GST,
an increase in the levels of oxido-nitrosative stress. The GR and iNOS enzymes, were also stimulated by curcumin.
etiology of the negative cellular effects of aging is unknown; Moreover, there was evidence for curcumin preventing the
however, several hypotheses exist including accumulation inhibition of Nrf2, NFĸB translocation and IĸB degradation,
of induced DNA damage, mitochondrial dysfunction, cellular all processes which are involved in regulating antioxidant
aging, and oxidative stress (López-Otín et al., 2013). ROS activity in the body. Oxidative or nitrosative stressors
has been highlighted as an important role player in DNA (e.g. rotenone, paraquat exposure) inhibit Nrf2 and stimulate
damage leading to aging. translocation of NFĸB in the cytosol and degradation of IĸB,
18 Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

indicated by the low


plasma curcumin levels
with 1 of the trials
reporting approximately
7.32 ng/ml (Ringman
et al., 2012). Therefore,
the positive findings
from animal and cellular
models may not be
directly translatable to
humans, and this has
led to research on alter-
native formulations of
curcumin to improve
its bioavailability (Chen
et al., 2018; Squillaro
et al., 2018). Other
clinical trials showed
70- and 180-fold in-
creases in curcumin
plasma when using
micelles as a novel
carrier system com-
pared to free curcumin
(Schiborr et al., 2014;
Fig. 1. The Proposed Antioxidant Effects of Curcumin Against DNA damage, and Lipid and Protein Oxidation. This schematic
diagram summarizes the main findings of the 64 articles summarized in this review. The arrow with a plus sign above indicates
Kocher et al., 2015).
stimulation while the T-shaped line indicates inhibition. The cross indicates protein degradation while the dashed lines highlight
processes which cause adverse effects to the cells including cellular dysfunction, damage and death. The exact cellular signal-
ing targets for curcumin have yet to be identified. GR: Glutathione reductase; GPx: Glutathione Peroxidase; H2O2: hydrogen
DISCUSSION AND
peroxide; HNE: 4-hydroxy-2-nonenal; IĸB: inhibitors-of-kappa B; MDA: malondialdehyde; NFĸB: nuclear-factor κB; NO: nitric CONCLUSION
oxide; NO2: nitrogen dioxide; N2O3: dinitrogen trioxide; Nrf2: nuclear factor erythroid 2–related factor 2; O2−: superoxide anion;
There are a number of
O2−2: peroxide; OH: hydroxyl radical; PN: peroxynitrate; SOD: superoxide dismutase.
completed or ongoing
clinical trials using cur-
which in turn suppresses antioxidant enzyme activity and cumin or its formulations and for many of these positive out-
enables unregulated production of ROS and RNS. Curcumin comes have been observed (NIH, 2018). However, there is
treatment, alternatively, could promote Nrf2 activation, and also some concern that curcumin and derivatives thereof
inhibit NFĸB translocation and IĸB degradation. Therefore, cur- are highly reactive, and have poor structural stability leading
cumin could stimulate the activities of antioxidant enzymes, to rapid metabolism and excretion. These issues may give
and thereby reduce the levels of oxidized proteins and lipid rise to false signals in drug screening tests and have led to
radicals to achieve oxidative homeostasis in cells. concerns regarding the efficacy of curcumin as a potential
therapeutic (Baker, 2016; Nelson et al., 2017).
In the 64 articles reviewed here, majority of the studies
reported in Tables 1 to 4 have produced promising results
CLINICAL TRIALS
for curcumin as an antioxidant therapy in NDDs and oxido-
According to clinicaltrials.gov, there are 13 ongoing and com- nitrosative stress. These protective effects have been observed
pleted clinical trials involving curcumin treatment and NDDs, for both animal and cellular models of these disorders. Curcu-
including AD, mild cognitive impairment and amyloidosis min was shown to be effective across a wide range of different
(https://clinicaltrials.gov). Currently, only 2 of the 13 clinical concentrations, when used before or after induction of oxidative
trials have been published in peer-reviewed journals (Baum stress, across different cell types and for various toxins. How-
et al., 2008; Ringman et al., 2012). In 1 of these clinical trials, ever, a major conflicting issue when analyzing the articles is
curcumin capsules or powder was ingested daily by 27 patients determination of the “protective” effects of antioxidant agents
with mild cognitive impairment for 6 months. No change in and enzymes in response to oxidative stress and curcumin.
levels of isoprostane and no improvement in cognitive decline Some studies report increased levels of SOD and reduced
at 1 and 4 g/day curcumin dosages were observed (Baum catalase and GSH levels, while others showed the converse,
et al., 2008). In the second study, no difference in iso- however, all of these studies concluded that curcumin was pro-
prostane, amyloid-β and tau plasma levels, at both 2- and tective. Furthermore, the use of a variety of toxins to elicit the
4-g/day dosages of curcumin were reported in a 24-week trial same disease makes comparing analyses between articles
of AD patients (Ringman et al., 2012). A possible reason for challenging, as it is uncertain whether the toxin is eliciting the
these findings could be due to the poor absorption of curcumin relevant disease pathways.
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 19

This review article is limited by the fact that only original Frautschy SA. (2008) Curcumin structure-function, bioavailability,
and efficacy in models of neuroinflammation and Alzheimer's disease.
journal articles in English were included, and only 3 online
J Pharmacol Exp Ther 326:196-208.
search databases, with a specific set of keywords, were Benn T, Halfpenny C, Scolding N. (2001) Glial cells as targets for cyto-
used. As a consequence of these filtering steps, relevant toxic immune mediators. Glia 36:200-211.
research articles may not have been retrieved. Bharath S, Hsu M, Kaur D, Rajagopalan S, Andersen JK. (2002)
In conclusion, curcumin is a promising agent for antioxida- Glutathione, iron and Parkinson's disease. Biochem Pharmacol
64:1037-1048.
tion (Fig. 1). However, further targeted experiments on curcu-
Bott LC, Badders NM, Chen K, Harmison GG, Bautista E, Shih CC,
min are needed to identify its exact molecular targets and the Katsuno M, Sobue G, Taylor JP, Dantuma NP, Fischbeck KH, Rinaldi
various pathways it is involved in before it can be used in the C. (2016) A small-molecule Nrf1 and Nrf2 activator mitigates polyglu-
design of novel therapeutics. tamine toxicity in spinal and bulbar muscular atrophy. Hum Mol Genet
25:1979-1989.
Braak E, Griffing K, Arai K, Bohl J, Bratzke H, Braak H (1999) Neuro-
pathology of Alzheimer's disease: what is new since A. Alzheimer?
CONFLICTS OF INTEREST Eur Arch Psychiatry Clin Neurosci 249 Suppl:14–22 Available at:
All authors declare no conflict of interest. http://link.springer.com/10.1007/PL00014168.
Canales-Aguirre AA, Gomez-Pinedo UA, Luquin S, Ramírez-Herrera MA,
Mendoza-Magaña ML, Feria-Velasco A. (2012) Curcumin protects
ACKNOWLEDGMENTS against the oxidative damage induced by the pesticide parathion in
the hippocampus of the rat brain. Nutr Neurosci 15:62-69.
We would like to acknowledge Prof Helena Kuivaniemi Chang C, Chen H, Yü G, Peng C. (2014) Curcumin-protected PC12 cells
(Stellenbosch University) for assisting in setting up the litera- against glutamate-induced oxidative toxicity. Food Technol Biotechnol
ture search and critically reviewing the manuscript. This work 52:468-478.
is based on the research supported wholly / in part by the Chen M, Du Z-Y, Zheng X, Li D-L, Zhou R-P, Zhang K. (2018) Use
of curcumin in diagnosis, prevention, and treatment of Alzheimer's
National Research Foundation of South Africa (Grant disease. Neural Regen Res 13:742-752 Available at: http://www.
Numbers: 111958, 106052, 96072), the South African nrronline.org/text.asp?2018/13/4/742/230303.
Medical Research Council (Self-Initiated Research Grant), Chen Q, Prior M, Dargusch R, Roberts A, Riek R, Chiruta C, Akaishi T,
and Stellenbosch University. WH is supported by the Faculty Abe K, Maher P, Schubert D. (2011) A novel neurotrophic drug for
cognitive enhancement and Alzheimer's disease. PLoS One 6e27865.
of Medicine and Health Sciences, Stellenbosch University.
Chen J, Tang XQ, Zhi JL, Cui Y, Yu HM, Tang EH, Sun SN, Feng JQ,
Chen PX. (2006) Curcumin protects PC12 cells against 1-methyl-4-
REFERENCES phenylpyridinium ion-induced apoptosis by bcl-2-mitochondria-ROS-
iNOS pathway*. Apoptosis 11:943-953.
Agrawal SS, Gullaiya S, Dubey V, Singh V, Kumar A, Nagar A, Tiwari P. Chen H, Xu D, Tan G, Cai W, Zhang G, Cui W, Wang J, Long C, Sun Y,
(2012) Neurodegenerative shielding by curcumin and its derivatives Yu P. (2015) A potent multi-functional neuroprotective derivative of
on brain lesions induced by 6-OHDA model of Parkinson's disease tetramethylpyrazine. J Mol Neurosci 56:977-987.
in albino Wistar rats. Cardiovasc Psychiatry Neurol 2012:1-8. Chhunchha B, Fatma N, Kubo E, Rai P, Singh SP, Singh DP. (2013)
Agrawal R, Mishra B, Tyagi E, Nath C, Shukla R. (2010) Effect of curcu- Curcumin abates hypoxia-induced oxidative stress based-ER stress-
min on brain insulin receptors and memory functions in STZ (ICV) mediated cell death in mouse hippocampal cells (HT22) by controlling
induced dementia model of rat. Pharmacol Res 61:247-252. Prdx6 and NF-κB regulation. Am J Physiol Cell Physiol 304:636-655.
Albrecht P, Lewerenz J, Dittmer S, Noack R, Maher P, Methner A (2010) Cui Q, Li XIN, Zhu H. (2016) Curcumin ameliorates dopaminergic neuro-
Mechanisms of oxidative glutamate toxicity: the glutamate/cystine nal oxidative damage via activation of the Akt/Nrf2 pathway. Mol Med
antiporter system xc as a neuroprotective drug target. CNS Neurol Rep 13:1381-1388.
Disord Targets (formerly Curr Drug Targets-CNS Neurol Disord Daverey A, Agrawal SK. (2016) Curcumin alleviates oxidative stress and
9:373–382. mitochondrial dysfunction in astrocytes. Neuroscience 333:92-103.
Alzheimer Association . (2015) 2015 Alzheimer's disease facts and Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB,
figures. Alzheimers Dement 11:332-384 Available at: https://doi.org/ Abbruzzese JL, Ng CS, Badmaev V, Kurzrock R. (2008) Phase II trial
10.1016/j.jalz.2015.02.003. of curcumin in patients with advanced pancreatic cancer. Clin Cancer
Amara F, Berbenni M, Fragni M, Leoni G, Viggiani S, Ippolito VM, Larocca Res 14:4491-4500.
M, Rossano R, Alberghina L, Riccio P, Colangelo AM (2015) Neuro- Dodson MW, Guo M. (2007) Pink1, Parkin, DJ-1 and mitochondrial dys-
protection by cocktails of dietary antioxidants under conditions of function in Parkinson's disease. Curr Opin Neurobiol 17:331-337.
nerve growth factor deprivation. Oxid Med Cell Longev:1–15. Finkel T, Holbrook NJ. (2000) Oxidants, oxidative stress and the biology
Arun S, Liu L, Donmez G. (2016) Mitochondrial biology and neurological of ageing. Nature 408:239-247.
diseases. Curr Neuropharmacol 14:143-154. Frautschy S, Hu W, Kim P, Miller S, Chu T, Harris-White G, Cole GM.
Baker M. (2016) Chemists warn against deceptive molecules: spice (2001) Phenolic anti-inflammatory antioxidant reversal of Aβ-induced
extract curcumin dupes assays and leads some drug hunters astray. cognitive deficits and neuropathology. Neurobiol Aging 22:993-1005.
Nature 541:144-145. Fu W, Wang H, Ren X, Yu H, Lei Y, Chen Q. (2017) Neuroprotective
Banji OJF, Banji D, Ch K. (2014) Curcumin and hesperidin improve cogni- effect of three caffeic acid derivatives via ameliorate oxidative
tion by suppressing mitochondrial dysfunction and apoptosis induced stress and enhance PKA/CREB signaling pathway. Behav Brain
by D-galactose in rat brain. Food Chem Toxicol 74:51-59. Res 328:81-86.
Banji D, Banji OJF, Dasaroju S, Kumar K. (2013) Curcumin and piperine Fu X Yan, Yang M Feng, Cao M Zhi, Li D Wei, Yang X Yi, Sun J Yi, Zhang
abrogate lipid and protein oxidation induced by D-galactose in rat Z Yong, Mao L Lei, Zhang S, Wang F Ze, Zhang F, Fan C Dong, Sun
brain. Brain Res 1515:1-11. B Liang. (2016) Strategy to suppress oxidative damage-induced neuro-
Baum L, Lam C, Cheung S, Kwok T, Lui V, Tsoh J, Lam L, Leung V, Hui toxicity in PC12 cells by curcumin: the role of ROS-mediated DNA
E, Ng C, Woo J. (2008) Six-month randomized, placebo-controlled, damage and the MAPK and AKT pathways. Mol Neurobiol 53:369-378.
double-blind, pilot clinical trial of curcumin in patients with Alzheimer Gebicki JM. (2016) Oxidative stress, free radicals and protein peroxides.
disease. J Clin Psychopharmacol 28:110-113. Arch Biochem Biophys 595:33-39.
Begum AN, Jones MR, Lim GP, Morihara T, Kim P, Heath DD, Rock CL, Harish G, Venkateshappa C, Mythri RB, Dubey SK, Mishra K, Singh N,
Pruitt MA, Yang F, Hudspeth B, Hu S, Faull KF, Teter B, Cole GM, Vali S, Bharath MMS. (2010) Bioconjugates of curcumin display
20 Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21

improved protection against glutathione depletion mediated oxidative Liu Z, Yu Y, Li X, Ross CA, Smith WW. (2011) Curcumin protects against
stress in a dopaminergic neuronal cell line: implications for Parkinson's A53T alpha-synuclein-induced toxicity in a PC12 inducible cell
disease. Bioorganic Med Chem 18:2631-2638. model for parkinsonism. Pharmacol Res 63:439-444.
Hayyan M, Hashim MA, AlNashef IM. (2016) Superoxide ion: generation Lobo V, Patil A, Phatak A, Chandra N. (2010) Free radicals, antioxidants
and chemical implications. Chem Rev 116:3029-3085. and functional foods: impact on human health. Pharmacogn Rev
Hosseinzadeh S, Roshan VD, Mahjoub S. (2013) Continuous exercise 4:118-126.
training and curcumin attenuate changes in brain-derived neuro- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. (2013)
trophic factor and oxidative stress induced by lead acetate in the The hallmarks of aging. Cell 153.
hippocampus of male rats. Pharm Biol 51:240-245. Lunn MR, Wang CH (2008) Spinal muscular atrophy. Semin 2120
Huang H, Lin C, Liu W, Jiang R, Jiang Z. (2011) Dual effects of curcumin www.thelancet.com 371 Available at: https://s3.amazonaws.com/
on neuronal oxidative stress in the presence of Cu(II). Food Chem academia.edu.documents/42185862/Spinal_muscular_
Toxicol 49:1578-1583. atrophy20160205-7989-ndai1.pdf?AWSAccessKeyId=
Ishrat T, Hoda N, Khan MB, Yousuf S, Ahmad M, Moshahid M, Ahmad A, AKIAIWOWYYGZ2Y53UL3A&Expires=1515445551&Signature=y%
Islam F. (2009) Amelioration of cognitive deficits and neurodegenera- 252FBGfWTcgspJP3FZ%252B%252BBCRDDNynE%
tion by curcumin in rat model of sporadic dementia of Alzheimer's type 253D&response-content-disposition=inline%253B.
(SDAT). Eur Neuropsychopharmacol 19:636-647. Luo Y, Hattori A, Munoz J, Qin Z, Roth GS. (1999) Intrastriatal dopamine
Jangra A, Kwatra M, Singh T, Pant R, Kushwah P, Sharma Y, Saroha B, injection induces apoptosis through oxidation involved activation
Datusalia AK, Bezbaruah BK. (2016) Piperine augments the protec- of transcription factors AP-1 and NF-κB in rats. Mol Pharmacol
tive effect of curcumin against lipopolysaccharide-induced neurobeha- 56:254-264.
vioral and neurochemical deficits in mice. Inflammation 39:1025-1038. Manogaran E, Ramanathan M, Ramarao T. (2015) Neuroprotective
Jaroonwitchawan T, Chaicharoenaudomrung N, Namkaew J, Noisa P. effect of curcumin against cholesterol induced neuroinflammation
(2017) Curcumin attenuates paraquat-induced cell death in human neu- in in-vitro and in-vivo models. J Pharm Sci Res 7:189-196.
roblastoma cells through modulating oxidative stress and autophagy. Marchiani A, Mammi S, Siligardi G, Hussain R, Tessari I, Bubacco L,
Neurosci Lett 636:40-47. Delogu G, Fabbri D, Ruzza P. (2013) Small molecules interacting
Jayaraj RL, Elangovan N, Manigandan K, Singh S, Shukla S. (2014) with a-synuclein: antiaggregating and cytoprotective properties.
CNB-001 a novel curcumin derivative, guards dopamine neurons in Amino Acids 45:327-338.
MPTP model of Parkinson's disease. Biomed Res Int 2014:1-11. Mishra S, Mishra M, Seth P, Sharma SK. (2011) Tetrahydrocurcumin
Jayaraj RL, Tamilselvam K. (2013) Neuroprotective effect of CNB-001, a confers protection against amyloid β-induced toxicity. Neuroreport
novel pyrazole derivative of curcumin on biochemical and apoptotic 22:23-27.
markers against rotenone-induced SK-N-SH cellular model of Montoya A, Price BH, Menear M, Lepage M. (2006) Brain imaging and
Parkinson's disease. J Mol Neurosci 51:863-870. cognitive dysfunctions in Huntington's disease. J Psychiatry Neurosci
Jiang T, Sun Q, Chen S. (2016) Oxidative stress: a major pathogenesis 31:21-29.
and potential therapeutic target of antioxidative agents in Parkinson's Morales I, Cerda-Troncoso C, Andrade V, Maccioni R. (2017) The natural
disease and Alzheimer's disease. Prog Neurobiol 147:1-19. product curcumin as a potential coadjuvant in Alzheimer's treatment.
Jiang H, Tian X, Guo Y, Duan W, Bu H, Li C. (2011) Activation of nuclear J Alzheimers Dis 60:451-460.
factor erythroid 2-related factor 2 cytoprotective signaling by curcumin Motaghinejad M, Motevalian M, Fatima S, Faraji F, Mozaffari S. (2017)
protect primary spinal cord astrocytes against oxidative toxicity. Biol The neuroprotective effect of curcumin against nicotine-induced
Pharm Bull 34:1194-1197. neurotoxicity is mediated by CREB–BDNF signaling pathway.
Kassebaum NJ, et al. (2016) Global, regional, and national disability- Neurochem Res 42:2921-2932.
adjusted life-years (DALYs) for 315 diseases and injuries and healthy Mythri RB, Harish G, Dubey SK, Misra K, Srinivas Bharath MM.
life expectancy (HALE), 1990–2015: a systematic analysis for the (2011) Glutamoyl diester of the dietary polyphenol curcumin
Global Burden of Disease Study 2015. Lancet 388:1603-1658. offers improved protection against peroxynitrite-mediated
Khatri DK, Juvekar AR. (2016) Neuroprotective effect of curcumin as nitrosative stress and damage of brain mitochondria in vitro:
evinced by abrogation of rotenone-induced motor deficits, oxidative implications for Parkinson's disease. Mol Cell Biochem
and mitochondrial dysfunctions in mouse model of Parkinson's 347:135-143.
disease. Pharmacol Biochem Behav 150–151:39-47. Nazari QA, Mizuno K, Kume T, Takada-Takatori Y, Izumi Y, Akaike A.
Kocher A, Schiborr C, Behnam D, Frank J (2015) The oral bioavailability (2012) In vivo brain oxidative stress model induced by microinjection
of curcuminoids in healthy humans is markedly enhanced by micellar of sodium nitroprusside in mice. J Pharmacol Sci 120:105-111 Avail-
solubilisation but not further improved by simultaneous ingestion of able at: http://japanlinkcenter.org/DN/JST.JSTAGE/jphs/12143FP?
sesamin , ferulic acid , naringenin and xanthohumol. J Funct Foods lang=en&from=CrossRef&type=abstract.
14:183–191 Available at: https://doi.org/10.1016/j.jff.2015.01.045. Nazari QA, Takada-takatori Y, Hashimoto T, Imaizumi A. (2014) Potential
Köstel AS, Bora-Tatar G, Erdem-Yurter H. (2012) Spinal muscular protective effect of highly bioavailable curcumin on an oxidative
atrophy: an oxidative stress response counteracted with curcumin. stress model induced by microinjection of sodium nitroprusside in
Biomed Aging Pathol 2:61-66. mice brain. Food Funct 5:984-989.
Kumar A, Prakash A, Dogra S. (2011) Protective effect of curcumin Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. (2017)
(Curcuma longa) against d-galactose-induced senescence in mice. The essential medicinal chemistry of curcumin. J Med Chem
J Asian Nat Prod Res 13:42-55. 60:1620-1637.
Lees AJ, Hardy J, Revesz T. (2009) Parkinson's disease. Lancet NIH (2018) ClinicalTrials.gov. Available at: https://clinicaltrials.gov/
373:2055-2066. search?term=curcumin [Accessed November 9, 2018].
Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Oliveira AS, Sousa E, Vasconcelos MH, Pinto M. (2015) Curcumin: a
Testa G, Cacciatore F, Bonaduce D, Abete P. (2018) Oxidative stress, natural lead for potential new drug candidates. Curr Med Chem
aging, and diseases. Clin Interv Aging 13:757-772. 22:4196-4232.
Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. (2001) The Ortiz-Ortiz MA, Morán JM, Bravosanpedro JM, González-Polo RA,
curry spice curcumin reduces oxidative damage and amyloid Niso-Santano M, Anantharam V, Kanthasamy AG, Soler G, Fuentes
pathology in an Alzheimer transgenic mouse. J Neurosci JM. (2009) Curcumin enhances paraquat-induced apoptosis of
21:8370-8377. N27 mesencephalic cells via the generation of reactive oxygen
Lin M-S, Hung K-S, Chiu W-T, Sun Y-Y, Tsai S-H. (2011) Curcumin species. Neurotoxicology 30:1008-1018.
enhances neuronal survival in N-methyl-d-aspartic acid toxicity by Öz A, Çelik Ö. (2016) Curcumin inhibits oxidative stress-induced
inducing RANTES expression in astrocytes via PI-3K and MAPK TRPM2 channel activation, calcium ion entry and apoptosis values
signaling pathways. Prog Neuropsychopharmacol Biol Psychiatry in SH-SY5Y neuroblastoma cells: involvement of transfection pro-
35:931-938. cedure. Mol Membr Biol 33:76-88.
Shameemah Abrahams et al. / Neuroscience 406 (2019) 1–21 21

Panchal HD, Vranizan K, Lee CY, Ho J, Ngai J, Timiras PS. (2008) Early Squillaro T, Cimini A, Peluso G, Giordano A, Melone MAB. (2018) Nano-
anti-oxidative and anti-proliferative curcumin effects on neuroglioma delivery systems for encapsulation of dietary polyphenols: an experi-
cells suggest therapeutic targets. Neurochem Res 33:1701-1710. mental approach for neurodegenerative diseases and brain tumors.
Park JH, Jung JW, Ahn Y, Kwon HW. (2012) Neuroprotective properties Biochem Pharmacol 154:303-317.
of phytochemicals against paraquat-induced oxidative stress and Sumanont Y, Murakami Y, Tohda M, Vajragupta O, Watanabe H,
neurotoxicity in Drosophila melanogaster. Pestic Biochem Physiol Matsumoto K. (2007) Effects of manganese complexes of curcumin
104:118-125. and diacetylcurcumin on kainic acid-induced neurotoxic responses
Pulido-Moran M, Moreno-Fernandez J, Ramirez-Tortosa C, Ramirez- in the rat hippocampus. Biol Pharm Bull 30:1732-1739.
Tortosa M. (2016) Curcumin and health. Molecules 21:264. Sun CY, Qi SS, Zhou P, Cui HR, Chen SX, Dai KY, Tang ML. (2013) Neu-
Ramkumar M, Rajasankar S, Gobi VV, Dhanalakshmi C. (2017) Neuro- robiological and pharmacological validity of curcumin in ameliorating
protective effect of demethoxycurcumin, a natural derivative of curcu- memory performance of senescence-accelerated mice. Pharmacol
min on rotenone induced neurotoxicity in SH-SY 5Y neuroblastoma Biochem Behav 105:76-82.
cells. BMC Complement Altern Med 17:217-227. Suresh D, Srinivasan K. (2010) Tissue distribution & elimination of capsai-
Rastogi M, Ojha RP, Sagar C, Agrawal A, Dubey GP. (2014) Protective cin, piperine & curcumin following oral intake in rats. Indian J Med Res
effect of curcuminoids on age-related mitochondrial impairment in 131:682-691.
female Wistar rat brain. Biogerontology 15:21-31. Suzuki YJ, Carini M, Butterfield DA (2010) Protein Carbonylation. Antioxid
Reyes-Fermin ML, Gonzalez-Reyes S, Tarco-Alvarez NG, Hernandez- Redox Signal 12:323–325 Available at: http://www.liebertonline.com/
Nava M, Orozco-Ibarra M, Pedraza-Chaverri J. (2012) Neuroprotec- doi/abs/10.1089/ars.2009.2887.
tive effect of α-mangostin and curcumin against iodoacetate-induced The Huntington's Disease Collaborative Research. (1993) A novel gene
cell death. Nutr Neurosci 15:34-41. containing a trinucleotide repeat that is expanded and unstable on
Ringman JM, Frautschy SA, Teng E, Begum AN, Bardens J, Beigi M, Huntington's disease chromosomes. Cell 72:971-983.
Gylys KH, Badmaev V, Heath DD, Apostolova LG, Porter V. (2012) Ullah F, Liang A, Rangel A, Gyengesi E, Niedermayer G, Münch G.
Oral curcumin for Alzheimer's disease: tolerability and efficacy in a (2017) High bioavailability curcumin: an anti-inflammatory and neuro-
24-week randomized, double blind, placebo-controlled study. Alzhei- supportive bioactive nutrient for neurodegenerative diseases charac-
mers Res Ther 4:43 Available at: http://alzres.com/content/4/5/43. terized by chronic neuroinflammation. Arch Toxicol 91:1623-1634.
Ruberti F, Capsoni S, Comparini A, Di Daniel E, Franzot J, Gonfloni S, Van Meeteren ME, Hendriks JJA, Dijkstra CD, Van Tol EAF. (2004)
Rossi G, Berardi N, Cattaneo A. (2000) Phenotypic knockout of nerve Dietary compounds prevent oxidative damage and nitric oxide
growth factor in adult transgenic mice reveals severe deficits in basal production by cells involved in demyelinating disease. Biochem
forebrain cholinergic neurons, cell death in the spleen, and skeletal Pharmacol 67:967-975.
muscle dystrophy. J Neurosci 20:2589-2601. Vidoni C, Castiglioni A, Seca C, Secomandi E, Melone MAB, Isidoro C.
Samy DM, Ismail CA, Nassra RA, Zeitoun TM, Nomair AM. (2016) Down- (2016) Dopamine exacerbates mutant huntingtin toxicity via
stream modulation of extrinsic apoptotic pathway in streptozotocin- oxidative-mediated inhibition of autophagy in SH-SY5Y neuro-
induced Alzheimer's dementia in rats: erythropoietin versus curcumin. blastoma cells: beneficial effects of anti-oxidant therapeutics.
Eur J Pharmacol 770:52-60. Neurochem Int 101:132-143 Available at: https://doi.org/10.1016/j.
Sandhir R, Yadav A, Mehrotra A, Sunkaria A, Singh A, Sharma S. (2014) neuint.2016.11.003.
Curcumin nanoparticles attenuate neurochemical and neurobe- Wang MS, Boddapati S, Emadi S, Sierks MR. (2010) Curcumin reduces
havioral deficits in experimental model of Huntington's disease. α-synuclein induced cytotoxicity in Parkinson's disease cell model.
Neuromolecular Med 16:106-118. BMC Neurosci 11:57-66.
Sarkar B, Dhiman M, Mittal S, Mantha AK (2017) Curcumin revitalizes Weidinger A, Kozlov AV. (2015) Biological activities of reactive oxygen
amyloid beta (25–35)-induced and organophosphate pesticides and nitrogen species: oxidative stress versus signal transduction.
pestered neurotoxicity in SH-SY5Y and IMR-32 cells via activation Biomolecules 5:472-484.
of APE1 and Nrf2. Metab Brain Dis:1–17. WHO . (2006) Neurological disorders: public health challenges. World
Schiborr C, Kocher A, Behnam D, Jandasek J, Toelstede S. (2014) The Health Organization, 2006.
oral bioavailability of curcumin from micronized powder and liquid & Wolfe MS, editor. The molecular and cellular basis of neurodegenera-
micelles is significantly increased. Mol Nutr Food Res 58:516-527. tive diseases: underlying mechanisms. Academic Press.
Schmidt MM, Dringen R. (2009) Differential effects of iodoacetamide and Yang D, Li T, Liu Z, Arbez N, Yan J, Moran TH, Ross CA, Smith WW.
iodoacetate on glycolysis and glutathione metabolism of cultured (2012) Neurobiology of disease LRRK2 kinase activity mediates toxic
astrocytes. Front Neuroenergetics 1:1-10. interactions between genetic mutation and oxidative stress in a
Seo J, Leem Y, Lee K, Kim S, Lee J, Han P. (2010) Severe motor neuron Drosophila model: suppression by curcumin. Neurobiol Dis 47:385-392.
degeneration in the spinal cord of the Tg2576 mouse model of Alzheimer Yu S, Wang X, He X, Wang Y, Gao S, Ren L, Shi Y. (2016) Curcumin
disease. J Alzheimers Dis 21:263-276. exerts anti-inflammatory and antioxidative properties in 1-methyl-4-
Sharma C, Suhalka P, Sukhwal P, Jaiswal N, Bhatnagar M. (2014) phenylpyridinium ion (MPP +)-stimulated mesencephalic astrocytes
Curcumin attenuates neurotoxicity induced by fluoride: an in vivo by interference with TLR4 and downstream signaling pathway. Cell
evidence. Pharmacogn Mag 10:61-65. Stress Chaperones 21:697-705.
Shelat DY, Acharya SR. (2016) Neuroprotective activity of novel CUR- Zhang L, Wu C, Zhao S, Yuan D, Lian G, Wang X, Wang L, Yang J.
CA-THIONE and its oxidative stress study. Int J Pharm Pharm Sci (2010) Demethoxycurcumin, a natural derivative of curcumin attenuates
8:167-173. LPS-induced pro-inflammatory responses through down-regulation of
Shin HJ, Lee JY, Son E, Lee DH, Kim HJ, Kang SS, Cho GJ, Choi WS, intracellular ROS-related MAPK/NF-κB signaling pathways in N9 micro-
Roh GS. (2007) Curcumin attenuates the kainic acid-induced hippo- glia induced by lipopolysaccharide. Int Immunopharmacol 10:331-338
campal cell death in the mice. Neurosci Lett 416:49-54. Available at: https://doi.org/10.1016/j.intimp.2009.12.004.
Sood PK, Nahar U, Nehru B. (2011) Curcumin attenuates aluminum- Zhao X, Zhang L, Yu H, Sun Z, Lin X, Tan C, Lu R. (2011) Curcumin pro-
induced oxidative stress and mitochondrial dysfunction in rat brain. tects mouse neuroblastoma neuro-2A cells against hydrogen-
Neurotox Res 20:351-361. peroxide-induced oxidative stress. Food Chem 129:387-394.

(Received 27 November 2018, Accepted 13 February 2019)


(Available online 28 February 2019)

View publication stats

You might also like