ArticlePDF Available

Curcumin Attenuates Colistin-Induced Neurotoxicity in N2a Cells via Anti-inflammatory Activity, Suppression of Oxidative Stress, and Apoptosis

Authors:

Abstract and Figures

Neurotoxicity is an unwanted side-effect seen in patients receiving therapy with the “last-line” polymyxin antibiotics. This is the first study to show that colistin-induced neurotoxicity in neuroblastoma-2a (N2a) cells gives rise to an inflammatory response involving the IL-1β/p-IκB-α/NF-κB pathway. Pretreatment with curcumin at 5, 10, and 20 μM for 2 h prior to colistin (200 μM) exposure for 24 h, produced an anti-inflammatory effect by significantly down-regulating the expression of the pro-inflammatory mediators cyclooxygenase-2 (COX-2), phosphorylation of the inhibitor of nuclear factor-kappa B (NF-κB) (p-IκB)-α, and concomitantly NF-κB levels. Moreover, curcumin significantly decreased intracellular reactive oxygen species (ROS) production and increased the activities of the anti-ROS enzymes superoxide dismutase, catalase, and the intracellular levels of glutathione. Curcumin pretreatment also protected the cells from colistin-induced mitochondrial dysfunction, caspase activation, and subsequent apoptosis. Overall, our findings demonstrate for the first time, a potential role for curcumin for treating polymyxin-induced neurotoxicity through the modulation of NF-κB signaling and its potent anti-oxidative and anti-apoptotic effects.
This content is subject to copyright. Terms and conditions apply.
Curcumin Attenuates Colistin-Induced Neurotoxicity in N2a Cells
via Anti-inflammatory Activity, Suppression of Oxidative Stress,
and Apoptosis
Chongshan Dai
1
&Giuseppe D. Ciccotosto
2
&Roberto Cappai
2
&Shusheng Tang
1
&
Daowen Li
1
&Sanlei Xie
1
&Xilong Xiao
1
&Tony Velkov
3
Received: 20 May 2016 / Accepted: 30 October 2016
#Springer Science+Business Media New York 2016
Abstract Neurotoxicity is an unwanted side-effect seen in
patients receiving therapy with the Blast-line^polymyxin an-
tibiotics. This is the first study to show that colistin-induced
neurotoxicity in neuroblastoma-2a (N2a) cells gives rise to an
inflammatory response involving the IL-1β/p-IκB-α/NF-κB
pathway. Pretreatment with curcumin at 5, 10, and 20 μMfor
2 h prior to colistin (200 μM) exposure for 24 h, produced an
anti-inflammatory effect by significantly down-regulating
the expression of the pro-inflammatory mediators
cyclooxygenase-2 (COX-2), phosphorylation of the inhibitor
of nuclear factor-kappa B (NF-κB) (p-IκB)-α, and concomi-
tantly NF-κB levels. Moreover, curcumin significantly de-
creased intracellular reactive oxygen species (ROS) produc-
tion and increased the activities of the anti-ROS enzymes su-
peroxide dismutase, catalase, and the intracellular levels of
glutathione. Curcumin pretreatment also protected the cells
from colistin-induced mitochondrial dysfunction, caspase
activation, and subsequent apoptosis. Overall, our findings
demonstrate for the first time, a potential role for curcumin
for treating polymyxin-induced neurotoxicity through the
modulation of NF-κB signaling and its potent anti-oxidative
and anti-apoptotic effects.
Keywords Curcumin .Colistin .Neurotoxicity .
Inflammation .Oxidative stress .Mitochondrial dysfunction
Introduction
Polymyxins (colistin and polymyxin B) are increasingly being
used as last-line therapy to treat infections caused by problem-
atic multidrug-resistant (MDR) Gram-negative pathogens,
such as Acinetobacter baumannii,Pseudomonas aeruginosa,
and Klebsiella pneumoniae [1,2]. An unwanted side-effect of
polymyxin therapy is neurotoxicity [24]. Patients receiving
intravenous colistin methanesulfonate (CMS; the inactive pro-
drug of colistin) have been reported to present with neurolog-
ical symptoms such as confusion, dizziness, facial/peripheral
paraesthesia, vertigo, seizures, and less commonly fatal effects
including respiratory muscle weakness, apnoea, and ataxia
[36]. Clearly, the development of neuroprotective agents
for co-administration is pivotal to improve the clinical utility
of these very important last-line antibiotics.
Curcumin (diferuloylmethane, a yellow pigment in spice
turmeric) is a natural product polyphenol extracted from the
rhizome of Curcuma longa (Fig. 1a). Apart from its culinary
applications in Indian cooking, it is well known that curcumin
possesses many beneficial pharmacological properties such as
anti-tumor, anti-inflammatory, and anti-oxidant activities
[79]. The anti-inflammatory effects of curcumin are mediat-
ed through its ability to suppress pro-inflammatory cytokines
such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-
Electronic supplementary material The online version of this article
(doi:10.1007/s12035-016-0276-6) contains supplementary material,
which is available to authorized users.
*Xilong Xiao
xiaoxl@cau.edu.cn
*Tony Velkov
Tony.Velkov@monash.edu
1
College of Veterinary Medicine, China Agricultural University, 2
Yuanmingyuan West Road, Beijing 100193, PeoplesRepublicof
China
2
Department of Pathology, Bio21 Molecular Science and
Biotechnology Institute, The University of Melbourne,
Parkville, VIC, Australia
3
Drug Delivery, Disposition and Dynamics, Monash Institute of
Pharmaceutical Sciences, Monash University, 381 Royal Parade,
Parkville, VIC 3052, Australia
Mol Neurobiol
DOI 10.1007/s12035-016-0276-6
Mol Neurobiol
6, pro-inflammatory enzymes (e.g., cyclooxygenase 2 (COX-
2)) and inflammatory transcription factors such as NF-κB
[10]. Moreover, curcumin exerts a cyto-protective anti-oxi-
dant action by suppressing the accumulation of damaging
ROS and the subsequent cellular apoptosis [9,1113].
Given that inflammation plays a key role in many disease
states, curcumin has been investigated as a therapeutic agent
for cancer, cardiovascular, neurological, autoimmune, and
metabolic diseases [10,11]. Notably, curcumin can cross the
blood-brain barrier, indicating a potential application as a neu-
roprotective agent [10,12,14]. Clearly, curcuminsanti-
inflammatory and anti-oxidative properties hold a great deal
of potential for its clinical utility in a large number of chronic
disease states. The primary aim of the present study was to
investigate the neuroprotective pathways of curcumin against
colistin-induced neurotoxicity using a mouse neuroblastoma-
2a (N2a) cell culture model. We also explored the mechanisms
by which curcumin suppresses oxidative stress, mitochondrial
dysfunction, and apoptosis in N2a neuronalcells. The present-
ed findings are discussed in context of the clinical potential of
curcumin as a co-administered neuroprotective agent during
polymxyin therapy.
Materials and Methods
Chemicals
Colistin (sulfate, 20,400 units/mg) was obtained from
Zhejiang Shenghua Biology Co., Ltd. (Zhengjiang, China).
Curcumin and bisdemethoxycurcumin (BDMC; both purity,
98 %) and was purchased from Aladdin Reagent Co., Ltd.
(Shanghai, China). 3-(4,5-dimetylthiazol-2-yl)-2, 5-
diphenyltetrazolium bromide (MTT), dimethyl sulfoxide
(DMSO), and sodium dodecylsulfonate (SDS) were pur-
chased from AMRESCO Inc. (Solon, OH, USA).
Rhodamine (Rh) 123 and 2,7-dichlorofluorescensin diacetate
(DCFH-DA) were purchased from Beyotime (Haimen,
China). Dulbeccos modified Eaglesmedium(DMEM)and
fetal bovine serum (FBS) were obtained from Life
Technologies Corporation (Grand Island, NY, USA). All other
reagents were of highest analytical grade available.
Cell Culture
The mouse N2a cells (ATCC CCL-131)wereculturedin
DMEM medium supplemented with 10 % (v/v) FBS, 110 mg/
L sodium pyruvate, 100 units/mL penicillin, and 100 μg/mL
streptomycin (Beyotime, Haimen, China) at 37 °C in 5 %
CO
2
. The media were changed once per day.
Mouse primary cortical neurons were prepared from C57/
Bl6 embryonic day 14 mice under sterile conditions using
procedures as previously described and approved by the
Melbourne University Animal Ethics Committee [15].
Briefly, embryonic day 14 C57/BL6 mice cortices were re-
moved, dissected free of meninges, and dissociated in
0.025 % (w/v) trypsin in Krebs buffer. The dissociated cells
were triturated using a filter-plugged fine pipette tip, pelleted,
resuspended in plating medium (minimum Eaglesmedium
containing 10 % fetal calf serum and 5 % horse serum), and
counted. Cortical neuronal cells were seeded at 150,000 cells/
well onto a poly-D-lysine-coated 48-well plate in plating me-
dium for 2 h then replaced with freshly prepared neurobasal
medium containing B27 supplements, gentamicin, and
0.5 mM glutamine (all tissue culture reagents were purchased
from Invitrogen (Australia) unless otherwise stated). The neu-
ronal cells were allowed to mature for 7 days in culture before
commencing treatment using freshly prepared primary culture
media (neurobasal medium plus B27 supplements (minus an-
ti-oxidants). All cultures were maintained in an incubator set
at 37 °C with 5 % CO
2
. This method resulted in cultures
highly enriched for neurons (>95 % purity) with minimal as-
trocyte and microglial contamination. The accumulation of
colistin in primary mouse cortical neurons was visualized
using an anti-polymyxin B mouse IgM antibody (Thermo
Fisher Scientific, Rockford, USA) as previously described
with minor modifications to the protocol [16,17]. Briefly,
the mAb was diluted to 1:500 and incubated with the
colistin-treated cells overnight at 4 °C. The cells were washed
and incubated with M.O. M. biotinylated anti-mouse second-
ary link (Vector Labs, CA, USA) for 10 min, followed by
incubation with an AlexaFluor647 Streptavidin conjugate at
a 1:500 dilution (Life Technologies, VIC, Australia).
Measurement of Cell Viability
Cell viability was measured using the MTT assay. Briefly, N2a
cells (1 × 10
4
) were seeded into 96-well tissue culture plates.
After culture for 12 h, cells were pretreated with different
doses of colistin (50800 μM). After 24 h, the medium was
discarded and replaced with 100 μL serum-free DMEM
Fig. 1 Protective effects of curcumin and bisdemethoxycurcumin on
colistin-induced cytotoxicity in N2a neuronal cells. aThe chemical struc-
tures of curcumin and bisdemethoxycurcumin. bDose-dependent toxicity
of colistin on N2a cell viability. c,dImpact of curcumin (Cur) and bis-
demethoxycurcumin (BDMC) pretreatment for 2 h (5, 10, and 20 μM) on
colistin-induced (200 μM) cytotoxicity in N2a cells (24 h incubation). e,f
Etoposide (Eto50;50μM) was used as the positive control treatment. g
The neuroprotective effect of curcumin pretreatment (5, 10, and 20 μM
for 2 h) in mouse primary cortical neurons against colistin-induced
(200 μM) cell death. The cell viability data were normalized and calcu-
lated as a percentage of untreated vehicle control values. hConfocal
fluorescence microscopy images of colistin-treated mouse primary corti-
cal neurons stained with anti-polymyxin monoclonal antibody (green
channel). All cell viability data shown represent as the mean ± SD from
five independent experiments. *p<0.05;**p<0.01compared with the
untreated control;
#
p<0.05;
##
p<0.01compared with the colistin or
etoposide treatment
Mol Neurobiol
containing 10 μL MTT (5 mg/mL) and the cells were incu-
bated for 4 h at 37 °C. Finally, the medium was discarded and
100 μL DMSO was added. After incubation for 20 min at
room temperature, the absorbance was read at 570 nm in a
micro-plate reader (Molecular Devices, Sunnyvale, CA,
USA). To assess the cyto-protective effects, cells were
pretreated with curcumin or BDMC (5, 10, or 20 μM) for
2 h. Following 2 h, the medium containing curcumin or
BDMC was discarded and the cells were washed with cold
PBS and incubated with colistin (200 μM) for an additional
24 h and then cell viability was assessed. As a positive control,
N2a cells were treated with etoposide (50 μM) for 24 h with or
without curcumin (or BDMC) pretreatment for 2 h at 37 °C.
Mouse primary cortical neurons were pretreated with
curcumin (5, 10, and 20 μM) or vehicle (DMSO) for 2 h then
media was removed and fresh NB media containing colistin
(200 μM) or vehicle (PBS) was added to the wells and incu-
bated for 24 h. At the conclusion of the experiment, MTT
reagent was added to the treated cultures for 3 h at 37 °C then
culture media removed and formazan by product fully dis-
solved using DMSO. A 100-μL aliquot of the MTT/DMSO
was transferred to a 96-well clear-walled plate and the absor-
bance measured at 570 nm in a FLUOstar Omega plate reader
(BMG Labtech, Vic, Australia). The data were normalized and
calculated as a percentage of untreated vehicle control values.
Measurement of Apoptosis
The apoptosis assay was performed using an Annexin V-FITC
apoptosis detection kit according to the manufacturersproto-
col (Vazyme Biotech Co., Ltd., Nanjing, China). Epotoside
(50 μM) was used as positive control treatment. For the flow
cytometric analysis, cells were harvested with 0.25 % trypsin
without EDTA, washed twice with cold PBS, and resuspended
in 500 μL binding buffer supplied by the manufacturer. The
cells were then incubated with 5 μL annexin V-FITC (40 μg/
mL) and 5 μL propidium iodide (40 μg/mL) in the dark for
10 min. Analysis was performed using a BD FACSAria
flow cytometer (Becton Dickinson, San Jose, CA, USA) set
at an excitation wavelength of 488 nm and an emission wave-
length of 605 nm.
Measurement of Intracellular ROS Generation
Intracellular ROS production was measured using the ROS-
specific fluorescent dye 2,7-dichlorofluorescein diacetate
(DCFH-DA) according to the manufacturers protocol
(Beyotime, Haimen, China). N2a cells were plated into 12-
well plates at a density of 2 × 10
5
cells per well and pretreated
with curcumin at final concentrations of 5, 10, or 20 μMfor
2 h at 37 °C and washed with cold PBS. Cells were then
treated with colistin (200 μM) for 24 h. The control cells were
treated with curcumin at 20 μM per se or the vehicle (0.1 %
DMSO in PBS). After treatment, DCFH-DA (10 μM) was
added into the medium for a further 30 min at 37 °C. The cells
were washed twice with PBS, harvested by trypsinization, and
analyzed for ROS production using a BD FACSAriaflow
cytometer set at an excitation and emission wavelength of 485
and 530 nm.
Measurement of Intracellular Superoxide Dismutase,
Catalase Activities, and Intracellular Glutathione Levels
The superoxide dismutase (SOD), catalase (CAT) activities,
and glutathione (GSH) levels were detected using specific
assay kits according to the manufacturers instructions
(Nanjing Jiancheng Co., Ltd., Nanjing, China). In brief, N2a
cells were plated into 6-well plates at a density of 5 × 10
5
cells
per well and pretreated with curcumin (5, 10, or 20 μM) at
37 °C for 2 h. After removing the medium-containing
curcumin, the cells were incubated colistin (200 μM) for
24 h. As a positive control cells were treated with etoposide
(50 μM) for 24 h. The negative control cells were treated with
curcumin (20 μM) or the vehicle (0.1 % DMSO in PBS). Cells
were washed with cold PBS and lysed using the cell lysis
buffer provided by the manufacturer. The cell lysates were
centrifuged at 14,000×gfor 10 min at 4 °C. Supernatants were
collected and assayed for SOD, CAT activities, and GSH
levels. Protein concentrations were quantified using the
BCA protein assay kit (Beyotime, Haimen, China).
Measurement of the Change in Mitochondrial Membrane
Potential (Δψm)
The Δψ
m
was detected using the fluorescent indicator JC-1
(Beyotime, Haimen, China). N2a cells were plated into 12-
well plates at a density of 2 × 10
5
cells per well and pretreated
with curcumin (20 μM) at 37 °C for 2 h, followed by treatment
with colistin (200 μM) for 24 h. Epotoside (50 μM) was used
as positive control treatment. After treatment, N2a cells were
incubated in DMEM containing 10 μM JC-1 at 37 °C for
15 min, washed with PBS, and observed under a fluorescence
microscope (Leica Microsystems, Wetzlar, Germany). A shift
of fluorescence from red to green represents a loss of ψ
m
.JC-1
red fluorescent emission (normal ψ
m
) was measured at
583 nm with an excitation wavelength of 525 nm, and JC-1
green fluorescence emission (loss of ψ
m
) was measured with
an excitation wavelength of 525 and emission wavelength of
530 nm. For quantitative analysis, at least 100 regions of in-
terest were selected in each group and the ratios between fluo-
rescence intensity in the green (low membrane potential) and
red (high membrane potential) channels in each region of
interest were calculated. An increase in the ratio was
interpreted as the loss of ψ
m
.
Mol Neurobiol
Measurement of Caspase-3/7 and Caspase-9 Activities
N2a cells (1.5 × 10
4
cells per well) were cultured in 96-well plates
and treated with curcumin (5, 10, or 20 μM) for 2 h at 37 °C.
After removing the medium-containing curcumin, the cells were
then incubated in media-containing colistin (200 μM)for24hat
37 °C. Epotoside (50 μM) was used as positive control treatment.
The caspase-3/7 and caspase-9 activities were determined using
the Caspase-Glo®-3/7 and caspase-9 assay kits according to the
manufacturers instructions (Promega Corp., Madison, USA).
The luminescence was measured using a micro-plate
luminometer (Molecular Devices, Sunnyvale, CA, USA).
Wester n Blo tting
Following treatment, N2a cells were harvested using a scraper
and lysed in 100 mM Tris-HCl, pH 7.4, 2 % (w/v)SDS,10%
(v/v) glycerol, and 1 mM PMSF. Cell lysates were centrifuged
at 14,000×gfor 15 min at 4 °C, and supernatants was subject-
ed to Western blotting analysis. The proteins were resolved
using 1012 % SDS-PAGE gels and then transferred to a
nitrocellulose membrane (Applygen Technologies Inc.
Beijing, China). The membranes were blocked with 5 % (w/
v) skim-milk powder dissolved in Tris-buffered saline and
Tween 20 (TBST) for 2 h, washed three times with TBST,
and then incubated overnight at 4 °C with rabbit polyclonal
antibodies against COX-2 (ABclonal Biotech Co. Ltd.,
Cambridge, USA), heme oxygenase (HO)-1, Bax, IL-1β,
COX-2, and nuclear factor erythroid 2-related factor 2 (Nrf2;
ProteinTech Group, Inc., USA), p-inhibitor of NF-κB(IκB)-α
(p-IκB-α; Ser32/36; Beijing Biosynthesis Biotechnology Co.,
Ltd., Beijing, China), NF-κB (p65), and glyceraldehyde 3-
phosphate dehydrogenase (GAPDH; all 1: 1000; Santa Cruz
Biotechnology, Inc., USA). After being washed three times
with TBST, the membranes were incubated for 1 h with the
corresponding species-specific horseradish peroxidase-
conjugated secondary antibodies (1:10,000). The blots were
visualized using Western luminescent detection kit (Vigorous
Biotechnology, Beijing, China). The protein load was normal-
ized to GAPDH and quantified using the ImageJ software
(National Institute of Mental Health, Bethesda, MD, USA).
Immunofluorescence Detection of NF-κBProtein
Expression
After treatment, N2a cells were fixed with 4 % paraformalde-
hyde at room temperature for 30 min. The cells were then
washed twice with PBS and then permeabilized with 1 %
Triton X-100 for 20 min. The cells were then incubated in
blocking buffer (2 % bovine serum albumin in PBS) for 2 h
at 37 °C. Cells were incubated with rabbit polyclonal antibod-
ies against NF-κB (1:200; Santa Cruz Biotechnology, Inc.,
USA) overnight at 4 °C, washed twicewith PBS, and followed
by co-incubation with Cy3-labeled goat anti-rabbit IgG sec-
ondary antibodies (Beyotime Institution of Biotechnology,
Haimen, China; both 1:1000) for 2 h at 37 °C. The cells were
then washed twice with PBS and incubated with 4,6-
diamidino-2-phenylindole nuclear stain for 5 min. The expres-
sion of NF-κB was observed under a fluorescence microscope
at 583 nm with an excitation wavelength of 525 nm.
Quantitative Reverse-Transcription (qRT) PCR
Examination
N2a cells were plated into 6-well culture plates (5 × 10
5
cells
per well) and pretreated with curcumin (5, 10, or 20 μM) at
37 °C for 2 h. After removing the medium-containing
curcumin, the cells were washed with cold PBS and incubated
with colistin (200 μM) for 24 h. The treated cells were har-
vested, and total RNA was isolated using the TRIzol extrac-
tion kit according to the manufacturers instructions
(Invitrogen Inc., Carlsbad, CA). The cDNA was synthesized
from ~2 μg of total RNA using the Prime Script RT-PCR kit
(Takara, Dalian, China). The PCR primers used were as fol-
lows: Nrf2: forward, 5-CAC ATT CCC AAA CAA GAT GC-
3,reverse,5-TCT TTT TCC AGC GAG GAG AT-3;HO-1:
forward, 5-CGT GCT CGA ATG AAC ACT CT-3,reverse,
5-GGA AGC TGA GAG TGA GGA CC-3; NF-κB: for-
ward, 5-CAC TGT CTG CCT CTC TCG TCT-3;reverse,
5-AAG GAT GTC TCC ACA CCA CTG-3;GAPDH:for-
ward, 5-ACA GTC CAT GCC ATC ACT GCC-3;reverse,
5-GCC TGC TTC ACC ACC TTC TTG-3. qRT-PCR were
performed using a Chromo 4instrument (Bio-Rad,
Hercules, CA), and the cycling conditions used were as fol-
lows: 95 °C for 10 min; 40 cycles of 95 °C for 15 s, 60 °C for
1 min, and 72 °C for 40 s. All reactions were conducted in
triplicate. The fold change in gene expression was calculated
using 2
ΔΔCt
after normalizing to the expression level of the
reference gene GAPDH.
Statistical Analysis
All data are expressed as the mean ± standard deviation (SD).
Data from the control and treatment groups were analyzed
with one-way analysis of variance, followed by the LSD post
hoc test using SPSS V13.0 (SPSS Inc., Chicago, USA). A p
value <0.05 was considered significant.
Results
Curcumin Attenuates Colistin-Induced Cytotoxicity
of N2a Neuronal Cells
Exposure of N2a cells to colistin produced a dose-dependent
decrease in cell viability (Fig. 1b). For the curcumin cyto-
Mol Neurobiol
protective studies, N2a cells were exposed to 200 μMcolistin
for 24 h, which reduced the cell viability to 47.2 % (p<0.01).
Pretreatment of the N2a cells with curcumin at 5, 10, and
20 μM for 2 h prior to colistin exposure significantly attenu-
ated colistin-induced cytotoxicity and increased the cell via-
bility to 54.7, 63.2, and 73.6 % (all p< 0.01) (Fig. 1c), respec-
tively. Likewise, pretreatment with 5, 10, and 20 μMofthe
curcumin analog BDMC (which accounts for ~17 % of the
composition of curcumin powder) for 2 h, increased N2a cell
viability to 51.4, 56.7 (p< 0.05), and 60.5 % (p<0.01),re-
spectively (Fig. 1d). These findings would suggest that
curcumin has a stronger cyto-protective activity compared
with its congener BDMC. Similarly, in the positive control
etoposide-treated (50 μM) control cells, curcumin showed a
stronger cyto-protective effect compared with BDMC
(Fig. 1e, f). The neuroprotective effect of curcumin (5, 10,
and 20 μM) was validated in cultures of mouse primary
cortical neurons treated with colistin (200 μM) (Fig. 1g).
Curcumin pretreatment at 20 μM effectively protected the
primary cortical neurons from colistin-induced cell death
(>50 % at 200 μM colistin); curcumin treatment per se had
no impact on cell viability. Furthermore, the accumulation of
colistin in the cortical cells was visualized using an anti-
polymyxin monoclonal antibody (Fig. 1h). The imaging
results revealed that colistin accumulates in both the cell
body and the dendrites.
Curcumin Attenuates Colistin-Induced Apoptosis in N2a
Cells
Exposure of N2a cells to 200 μMcolistinfor24hin-
duced apoptotic rates up to 46.4 % (p< 0.01) (Fig. 2a).
Pretreatment of the N2a cells with curcumin at 5, 10, and
20 μM for 2 h prior to colistin exposure decreased the
Fig. 2 Protective effect of curcumin against colistin-induced apoptosis in
N2a cells. aApoptosis of N2a cells were analyzed by flow cytometry
following annexin V-FITV/PI staining. Q1 necrosis cells, Q2 later apo-
ptotic cells, Q3 live cells, Q4 early apoptotic cells. Cells were pretreated
with curcumin for 2 h at 37 °C. After removing the medium-containing
curcumin, the cells were then incubated in media-containing colistin for
24 h at 37 °C. bEtoposide (Eto50;50μM) was used as the positive
control treatment. The data shown represent the mean ± SD, from three
independent experiments. **p<0.01compared with the untreated con-
trol;
##
p<0.01compared with the colistin or etoposide treatment
Mol Neurobiol
Mol Neurobiol
apoptotic rates to 42.2, 34.4 (p< 0.01), and 24.5 %
(p< 0.01) (Fig. 2a), respectively. In the positive etoposide
(50 μM) control cells, pretreatment with curcumin
(20 μM) decreased apoptotic rates to 22.6 vs 33.4 % (no
pretreatment) (Fig. 2b). Curcumin treatment per se
(20 μM) had no effect on the apoptotic rates and apoptotic
rates were essentially comparable with untreated control
cells.
Curcumin Attenuates Colistin-Induced Mitochondrial
Dysfunction and Caspase Activation
The neuroprotective effect of curcumin against colistin-
induced mitochondrial dysfunction was assessed using the
JC-1 MTP assay. Colistin treatment (200 μM) for 24 h in-
duced mitochondrial dysfunction in N2a cells seen as an in-
crease of green fluorescent JC-1 (JC-1 monomeric form); the
green/red fluorescence radio increased 3.12 (p<0.01),com-
pared with that in the control (Fig. 3a, b). This indicated the
loss of mitochondrial membrane potential (ψ
m
). Pretreatment
of the N2a cells with 20 μM curcumin for 2 h prior to colistin
exposure protected the N2a cellular ψ
m
against colistin-
induced perturbations, as evidenced by the decreases of the
green/red fluorescence radio (decreased to 0.72; p< 0.01),
compared with the colistin-alone group (Fig. 3a, b). Colistin
exposure at 200 μM for 24 h significantly (all p< 0.01) in-
creased the activities of caspase-9 and caspase-3 to about 2.4-
and 2.8-fold, respectively, compared with the untreated con-
trol cells. Curcumin pretreatment (20 μM), significantly (all
p< 0.01) down-regulated the activation of caspase-9 and
caspase-3 to about 1.5- and 1.7-fold, respectively, compared
with the colistin only treated cells (Fig. 3c, d). In the positive
control etoposide-treated (50 μM) cells, pretreatment with
curcumin (20 μM) significantly (p< 0.05 or 0.01) attenuated
the loss of ψ
m
and down-regulated the activation of caspase-9
and caspase-3 (Fig. 3ad).
Curcumin Attenuates Colistin-Induced Oxidative Stress
by Decreasing Intracellular ROS, Inducing SOD and CAT
Activities, and Increasing Intracellular GSH Levels
Pretreatment of the N2a cells with curcumin at 5, 10, and
20 μM for 2 h prior to colistin exposure significantly de-
creased the intracellular ROS levels from 227.6 % (colistin
only treatment, to 161.3, 134.6, and 117.8 % (all p<0.01),
respectively (Fig. 4a). Moreover, curcumin pretreatment at
10 and 20 μM significantly increased (p<0.05and
p< 0.01, respectively) the SOD and CAT activities and the
intracellular levels of GSH (Fig. 4bd). Similarly, in the
positive etoposide (50 μM) control cells, curcumin pretreat-
ment at 20 μM markedly increased the activities of SOD and
CAT and the intracellular levels of GSH (all p<0.05or
0.01) (Fig. 4bd). Curcumin treatment per se (20 μM) had
no effect on cellular ROS, SOD, and CAT activities and
GSH levels (Fig. 4).
Curcumin down-Regulates the Activation
of Colistin-Induced Pro-inflammatory Mediators,
Pro-apoptotic Proteins, and Nrf2/HO-1 Expression
The protein expression in N2a cells of key pro-inflammatory
mediators including p-IκB-α,NF-κB, IL-1β, COX-2, the pro-
apoptotic protein Bax, and the Nrf2/HO-1 pathway proteins
were examined using Western blotting. Following colistin ex-
posure (200 μM) for 24 h, the expression of p-IκB-α(~18.1-
fold), NF-κB (~16.3-fold), IL-1β(~8.1-fold), COX-2 (~14.7-
fold), Bax (~2.2-fold), Nrf2 (~1.6-fold), and HO-1 (~3.4-
fold), all significantly increased (all p<0.01),comparedwith
the untreated control cells (Fig. 5). Pretreatment of the N2a
cells with curcumin (5, 10, and 20 μM) prior to colistin expo-
sure significantly decreased (p< 0.05 or 0.01) the expression
of p-IκB-α,NF-κB, COX-2, and Bax, in a dose-dependent
manner (Fig. 5). Conversely, curcumin pretreatment increased
the expression of Nrf2 and HO-1 and had no effect on the
expression of IL-1β(Fig. 5). Interestingly, in the positive
etoposide (50 μM) control cells, curcumin pretreatment
(20 μM) decreased the expression of p-IκB-α,NF-κB,
COX-2, and IL-1β(all p< 0.05 or 0.01) and increased the
expression of Nrf2 and HO-1 (Fig. S1). Furthermore, NF-κB
expression in N2a cells in response to colistin exposure de-
tected via immunofluorescence (Fig. 6), showed that
curcumin down-regulates the colistin- or etoposide-induced
expression of NF-κB(Fig.5).
Curcumin Increases Nrf2 and HO-1 mRNA Expression
and Decreases NF-kB mRNA Expression
Exposure of N2a cells to colistin (200 μM) for 24 h signifi-
cantly (all p< 0.05 to <0.01) increased the messenger RNA
(mRNA) expression of Nrf2, OH-1, and NF-κB (Fig. S2).
Fig. 3 Curcumin attenuates colistin-induced mitochondrial dysfunction
and caspase activation. aN2a cells were plated into 12-well plates at a
density of 2 × 10
5
cells per well and pretreated with curcumin at final
concentrations of 20 μM(Cur20)at3Cfor2h(n= 3). The cells were
treated with or without colistin at 200 μM for additional 24 h. Etoposide
(Eto50;50μM) was used as the positive control treatment. The change in
mitochondrial membrane potential (MTP) was evaluated using the cat-
ionic fluorescent indicator JC-1. The JC-1-aggregate form, indicating
normal MTP function, appears red. The JC-1 monomeric form, indicating
disrupted MTP, appears green. Magnification, ×40. bQuantitative anal-
ysis of the confocal data are presented as the ratio between fluorescence
intensityinthegreen (low membrane potential) and red (high membrane
potential) channels. An increase in the ratio was interpreted as the loss of
ψ
m
.c,dCaspase-9 and caspase-3 activity in N2a cells in response to
colistin and curcumin pretreatment were examined using ELISA. Values
are presented as the mean ± SD, from three independent experiments.
*p<0.05;**p<0.01compared with the untreated control;
#
p<0.05;
##
p<0.01compared with the colistin or etoposide treatment
Mol Neurobiol
Pretreatment with curcumin (20 μM) for 2 h markedly de-
creased the expression of NF-κB mRNA and significantly
increased the expression of Nrf2 and OH-1 mRNA, in a
dose-dependent manner (Fig. S2). The expression levels of
the reference gene GAPDH did not change under the condi-
tions used.
Discussion
Neurotoxicity is a major, albeit poorly understood side-effect
associated with polymyxin therapy [3,4,6,18]. We have
previously shown using an in vitro N2a cell culture model that
colistin-induced neurotoxicity involves apoptosis,
Fig. 4 Curcumin protects N2a
cells against colistin-induced oxi-
dative stress. aCellular ROS
levels were detected using flow
cytometry following staining of
N2a cells with the ROS-sensitive
dye 2,7-dichlorofluorescein
diacetate. bdThe impact of
curcumin pretreatment (5, 10, and
20 μM) for 2 h on cellular super-
oxide dismutase (SOD), catalase
(CAT) activities, and glutathione
(GSH) levels in N2a cells treated
with colistin (200 μM for 24 h).
Etoposide (Eto50;50μM) was
used as the positive control treat-
ment. The data shown represent
the mean ± SD, from three inde-
pendent experiments.
**p<0.01compared with the
untreated control;
#
p<0.05;
##
p<0.01compared with the
colistin or etoposide treatment
Mol Neurobiol
mitochondrial dysfunction, generation of ROS, and autopha-
gy [19]. The discovery of neuroprotective agents for co-
administration during colistin therapy is paramount to prolong
the clinical utility of this important last-line lipopeptide anti-
biotic. The aim of the present study was to exploit our in vitro
N2a neuronal cell culture model of colistin-induced neurotox-
icity to study the neuroprotective mechanism of curcumin in
antagonizing pro-inflammatory cytokines by suppressing
NF-κB activation and signaling. We also explored the neuro-
protective molecular mechanisms by which curcumin sup-
press apoptosis, mitochondrial dysfunction, and ROS produc-
tion in the N2a neuronal cells.
Compositionally, curcumin (curry powder) is predominantly
diferuloylmethane (1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-
heptadiene-3,5 dione) and contains ~17 % bisdemethoxycurcu-
min (BDMC) and 3 % demethoxycurcumin (DMC) [2022].
Direct curcumin targets include COX-2, lipoxygenase, toll-like
receptor (TLR)4, focal adhesion kinase, glutathione, glycogen
synthase kinase (GSK)-3 β, phosphorylase-3 kinase, xanthine
oxidase, pp60 src tyrosine kinase, and ubiquitin isopeptidase
[2328]. The molecular targets indirectly regulated by curcumin
include the transcription factors NF-κB, hypoxia inducible factor
(HIF)-1a, activator protein (AP)-1, signal transducers and
activators of transcription protein (STAT)-3, peroxisome
proliferator-activated receptors, p53, anti-apoptotic proteins
(e.g., Bcl-2 and Bcl-xL), cell-cycle regulatory proteins (e.g.,
cyclins D1, E, and c-myc), and inflammatory mediators (e.g.,
IL-1 and IL-6) [10,25,29,30].
In our recent mechanistic toxicity study, we reported that
colistin-induced apoptosis in neuronal N2a cells is activated
via both the death receptor (extrinsic) and the mitochondrial
(intrinsic) pathways [19]. In the present report, we have shown
that colistin-induced apoptosis could be inhibited by pretreat-
ment of the N2a cells with 20 μMofcurcuminfor2h.
Specifically, curcumin pretreatment significantly attenuated the
loss of mitochondrial membrane potential (Fig. 3a) and down-
regulated the expression of the pro-apoptotic protein Bax (Fig. 5)
and the activation of caspase-9 and caspase-3 (Fig. 3b, c).
The nervous system is highly vulnerable to oxidative damage
due to its elevated oxygen demand and high polyunsaturated
fattyacidcontent[31]. We previously reported that colistin-
induced neurotoxicity involves mitochondrial dysfunction in
the mouse cerebral cortex and sciatic nerve tissues in mice in-
travenously injected with 15 mg kg
1
day
1
colistin sulfate for
7days[32]. In the present study, we found that colistin exposure
significantly increased intracellular ROS levels in N2a cells with
Fig. 5 Curcumin attenuates colistin-induced expression of pro-
inflammatory cytokines and pro-apoptotic protein and activates the
Nrf2/HO-1 pathway. Left panels, N2a cells treated with colistin
(200 μM for 24 h) with or without curcumin pretreatment (5, 10, and
20 μM for 2 h) were analyzed using western blotting for expression of p-
IκB-α,NF-κB, interleukin (IL)-1β, cyclooxygenase-2 (COX-2), nuclear
factor erythroid 2-related factor 2 (Nrf2; band indicated with asterisk),
heme oxygenase (HO)-1, and Bax. Glyceraldehyde 3-phosphate dehy-
drogenase (GAPDH) was used to demonstrate equal protein loading per
gel lane. Right panels, the relative protein expression levels were ana-
lyzed using ImageJ. Values are presented as mean ± SD, from three
independent experiments. *p<0.05;**p<0.01compared with the
untreated control;
#
p<0.05;
##
p<0.01compared with the colistin or
etoposide treatment
Mol Neurobiol
a concomitant decrease in the activity of the anti-oxidant en-
zymes SOD and CAT and decreased intracellular GSH levels
(Fig. 4). Taken together, these findings would suggest that co-
listin neurotoxicity not only induces ROS production directly, it
further decreases the neuronscapacity to breakdown oxygen
radicals, further exacerbating ROS-mediated oxidative stress.
One of the most remarkable pharmacological properties of
curcumin is its anti-oxidant activity, indeed it has been reported
that curcumin can mop up oxygen radicals even more effective-
ly than vitamin E [26]. In the present study, we found that
curcumin could not only inhibit colistin-induced ROS genera-
tion but also enhance the total anti-oxidant capacity in N2a cells
by up-regulating the activities of SOD and CAT and increasing
intracellular GSH levels (Fig. 4). The potent anti-oxidant activ-
ity of curcumin is closely related to its ability to activate Nrf2, a
transcription factor which mediates the expression of anti-
oxidant mediators such as CAT, SOD, and HO-1 [8,11].
Notably, curcumin has been shown to act as a neuroprotectant
against methylmercury and 6-hydroxydopamine-induced neu-
rotoxicity via the up-regulation of Nrf2 and HO-1 expression
[33,34]. Similarly in the present study, we found that curcumin
may counteract colistin-induced neurotoxicity by increasing the
expression of the anti-oxidant response factors Nrf2 and HO-1
(Fig. 5;Fig.S2).
Fig. 6 Immunofluorescence
analysis of NF-κBproteinex-
pression in N2a cells. N2a cells
were pretreated with curcumin
(Cur;20μM) or bisdemethoxy-
curcumin (BDMC;20μM) for
2 h, followed by incubation with
colistin (200 μM) for an addi-
tional 24 h (n= 3). The N2a cells
were stained with anti-NF-κB
antibody. The nucleus was stained
with DAPI dye (blue). Etoposide
(Eto;50μM) was used as the
positive control treatment. The
magnified region in the colistin
only condition shows the nuclear
NF-κB. Magnification, ×40
Mol Neurobiol
To the best of our knowledge, this is the first study to report
that colistin-induced neurotoxicity involves the inflammatory
response via IL-1βand NF-κB pathway signaling with sub-
sequent induction of the inflammatory enzyme COX-2
(Fig. 5). Notably, polymyxin B has been previously reported
to induce the section of IL-1βin bone marrow dendritic cells
and macrophages and induce the expression of tumor necrosis
factor (TNF)-αin human peripheral blood mononuclear cells
[35,36]. NF-κB plays a central role in the cellular response to
diverse array of inflammatory stimuli [37]. Notably, NF-κB
activation is known to regulate the expression of over
500 genes associated with inflammation, tumorgenesis,
cellular survival/proliferation, and chemoresistance [38].
Accordingly, inhibitors of NF-κB activation such as curcumin
hold a great deal of therapeutic potential for the treatment of
inflammatory diseases. Phase I clinical trials with human sub-
jects indicated that oral curcumin doses of 812 g/day had no
adverse effects [39,40]. NF-κB exists in an inactive state in
the cytoplasm as a heterotrimer consisting of two subunits and
an inhibitory IκB-αsubunit [41]. NF-κB activation and nu-
clear translocation involves IKK kinase-mediated phosphory-
lation of the inhibitory IκB-αsubunit, which is released from
the NF-κB complex and is subsequently degraded [37,41,
42]. Once activated, the NF-κB subunits translocate into the
nucleus and bind to the promoters of target genes encoding
pro-inflammatory cytokines (IL-1, IL-2, IL-6, TNF-α) and
pro-inflammatory enzymes including COX-2 and inducible
nitric oxide synthase [43] which exacerbate and perpetuate
the inflammatory response by initiating catabolic processes
[44]. It is well documented that the anti-inflammatory mode
of action of curcumin involves the down-regulation of NF-kB
signaling and the mediators of inflammation (e.g., COX-2) by
inhibiting the activity of IKK kinase [42,4547]. In the pres-
ent study, we found that curcumin can inhibit colistin-induced
expression of NF-κB and markedly decrease the expression of
p-IκB-αand COX-2; however, there was no effect on the
expression of IL-1β. It has been reported that HO-1 activation
inhibits the nuclear translocation of NF-κB[48]. Coincidently,
we observed that curcumin treatment significantly increased
the expression of HO-1 which may be partially responsible for
concomitant down-regulated expression of NF-κB.
Overall, our data suggest that curcumin attenuates colistin-
induced neurotoxicity in N2a cells by down-regulating NF-kB
and NF-kB-regulated genes involved in inflammation and ap-
optosis. These anti-inflammatory and anti-apoptotic neuropro-
tective effects are mediated partly via the down-regulation of
the phosphorylation of IκB-α, suppression of oxidative stress,
and mitochondrial-mediated apoptotic pathways (Fig. 7).
Although curcumin exhibits poor systemic availability when
administered via the oral route, various formulations for
intravenous use, including nanoparticles and liposomal
encapsulation have allowed for improved dosing and have
shown promise as anti-neoplastic agents [4951]. Notably,
available preclinical and phase I/II data suggest that curcumin
is well tolerated, and has a good safety profile [50]. This study
provides fundamental supportive data for the clinical applica-
tion of curcumin as a co-administered neuroprotective agent
during colistin therapy of MDR Gram-negative infections.
Acknowledgements This study was supported by Key Projects in the
National Science and Technology Pillar Program during the 12th Five-
Year Plan Period (2015BAD11B03) and Chinese Universities Scientific
Fund (award number 2015DY003). T. V. is supported by a research grant
from the National Institute of Allergy and Infectious Diseases of the
National Institutes of Health (R01 AI111965). T. V. is also supported by
the Australian National Health and Medical Research Council
(NHMRC).
Compliance with Ethical Standards
Conflict of Interest The authors declare that there are no conflicts of
interest.
References
1. Li J, NationRL, Turnidge JD, Milne RW, Coulthard K,Rayner CR,
Paterson DL (2006) Colistin: the re-emerging antibiotic for
multidrug-resistant gram-negative bacterial infections. Lancet
Infect Dis 6(9):589601. doi:10.1016/S1473-3099(06)70580-1
2. Nation RL, Li J, Cars O, Couet W, Dudley MN, Kaye KS, Mouton
JW, Paterson DL et al (2015) Framework for optimisation of the
clinical use of colistin and polymyxin B: the Prato polymyxin
Fig. 7 Schematic representation of the protective effects of curcumin on
colistin-induced oxidative stress, apoptosis, and inflammatory responses
in N2a neuronal cells
Mol Neurobiol
consensus. Lancet Infect Dis 15(2):225234. doi:10.1016/S1473-
3099(14)70850-3
3. Falagas ME, Kasiakou SK (2006) Toxicity of polymyxins: a sys-
tematic review of the evidence from old and recent studies. Crit
Care 10 (1). doi: 10.1186/cc3995
4. Wahby K, Chopra T, Chandrasekar P (2010) Intravenous and inha-
lational colistin-induced respiratory failure. Clin Infect Dis 50(6):
e38e40. doi:10.1086/650582
5. Weinstein L, Doan TL, Smith MA (2009) Neurotoxicity in patients
treated with intravenous polymyxin B: two case reports. Am J
Health Syst Pharm 66(4):345347. doi:10.2146/ajhp080065
6. Honore PM, Jacobs R, Lochy S, De Waele E, Van Gorp V, De Regt
J, Martens G, Joannes-Boyau O et al (2013) Acute respiratory mus-
cle weakness and apnea in a critically ill patient induced by colistin
neurotoxicity: key potential role of hemoadsorption elimination
during continuous venovenous hemofiltration. Int J Nephrol
Renovasc Dis 6:107111. doi:10.2147/IJNRD.S42791
7. Zhao XC, Zhang L, Yu HX, Sun Z, Lin XF, Tan C, Lu RR (2011)
Curcumin protects mouse neuroblastoma neuro-2A cells against
hydrogen-peroxide-induced oxidative stress. Food Chem 129(2):
387394. doi:10.1016/j.foodchem.2011.04.089
8. Sahin K, Orhan C, Tuzcu Z, Tuzcu M, Sahin N (2012) Curcumin
ameloriates heat stress via inhibition of oxidative stress and modu-
lation of Nrf2/HO-1 pathway in quail. Food Chem Toxicol 50(11):
40354041. doi:10.1016/j.fct.2012.08.029
9. Tokac M, Taner G, Aydin S, Ozkardes AB, Dundar HZ, Taslipinar
MY, Arikok AT, Kilic M et al (2013) Protective effects of curcumin
against oxidative stress parameters and DNA damage in the livers
and kidneys of rats with biliary obstruction.Food Chem Toxicol 61:
2835. doi:10.1016/j.fct.2013.01.015
10. Ghosh S, Banerjee S, Sil PC (2015) The beneficial role of curcumin
on inflammation; diabetes and neurodegenerative disease: a recent
update. Food Chem Toxicol 83:111124. doi:10.1016/j.
fct.2015.05.022
11. Carmona-Ramirez I, Santamaria A, Tobon-Velasco JC, Orozco-
Ibarra M, Gonzalez-Herrera IG, Pedraza-Chaverri J, Maldonado
PD (2013) Curcumin restores Nrf2 levels and prevents quinolinic
acid-induced neurotoxicity. J Nutr Biochem 24(1):1424.
doi:10.1016/j.jnutbio.2011.12.010
12. Garcia-Nino WR, Pedraza-Chaverri J (2014) Protective effect of
curcumin against heavy metals-induced liver damage. Food Chem
Toxicol 69:182201. doi:10.1016/j.fct.2014.04.016
13. Sharma C, Suhalka P, Sukhwal P, Jaiswal N, Bhatnagar M (2014)
Curcumin attenuates neurotoxicity induced by fluoride: an in vivo
evidence. Pharmacogn Mag 10(37):6165. doi:10.4103/0973-
1296.126663
14. Barta A, Janega P, Babal P, Murar E, Cebova M, Pechanova O
(2015) The effect of curcumin on liver fibrosis in the rat model of
microsurgical cholestasis. Food Funct 6(7):21872193.
doi:10.1039/c5fo00176e
15. Jana MK, Cappai R, Pham CL, Ciccotosto GD (2016) Membrane-
bound tetramer and trimer Abeta oligomeric species correlate with
toxicity towards cultured neurons. J Neurochem 136(3):594608.
doi:10.1111/jnc.13443
16. Yun B, Azad MA, Wang J, Nation RL, Thompson PE, Roberts
KD, Velkov T, Li J (2015) Imaging the distribution of poly-
myxins in the kidney. J Antimicrob Chemother 70(3):827829.
doi:10.1093/jac/dku441
17. Velkov T, Yun B, Schneider EK, Azad MA, Dolezal O, Morris FC,
Nation RL, Wang J et al (2016) A novel chemical biology approach
for mapping of polymyxin lipopeptide antibody binding epitopes.
ACS Infect Dis 2(5):341351. doi:10.1021/acsinfecdis.6b00031
18. Shrestha A, Soriano SM, Song M, Chihara S (2014) Intravenous
colistin-induced acute respiratory failure: a case report and a review
of literature. Int J Crit Illn Inj Sci 4(3):266270. doi:10.4103/2229-
5151.141487
19. Dai C, Tang S, Velkov T, Xiao X (2016) Colistin-induced apoptosis
of neuroblastoma-2a cells involves the generation of reactive oxy-
gen species, mitochondrial dysfunction, and autophagy. Mol
Neurobiol 53(7):4685-700. doi:10.1007/s12035-015-9396-7
20. Yodkeeree S, Chaiwangyen W, Garbisa S, Limtrakul P (2009)
Curcumin, demethoxycurcumin and bisdemethoxycurcumin differ-
entially inhibit cancer cell invasion through the down-regulation of
MMPs and uPA. J Nutr Biochem 20(2):8795. doi:10.1016/j.
jnutbio.2007.12.003
21. Sandur SK, Pandey MK, Sung B, Ahn KS, Murakami A, Sethi G,
Limtrakul P, Badmaev V et al (2007) Curcumin, demethoxycurcu-
min, bisdemethoxycurcumin, tetrahydrocurcumin and turmerones
differentially regulate anti-inflammatory and anti-proliferative re-
sponses through a ROS-independent mechanism. Carcinogenesis
28(8):17651773. doi:10.1093/carcin/bgm123
22. Huang MT, Ma W, Lu YP, Chang RL, Fisher C, Manchand PS,
Newmark HL, Conney AH (1995) Effects of curcumin, demetho-
xycurcumin, bisdemethoxycurcumin and tetrahydrocurcumin on
12-O-tetradecanoylphorbol-13-acetate-induced tumor promotion.
Carcinogenesis 16(10):24932497
23. Skrzypczak-Jankun E, Zhou K, McCabe NP, Selman SH, Jankun J
(2003) Structure of curcumin in complex with lipoxygenase and its
significance in cancer. Int J Mol Med 12(1):1724
24. Bharti AC, Donato N, Singh S, Aggarwal BB (2003) Curcumin
(diferuloylmethane) down-regulates the constitutive activation of nu-
clear factor-kappa B and IkappaBalpha kinase in human multiple my-
eloma cells, leading to suppression of proliferation and induction of
apoptosis. Blood 101(3):10531062. doi:10.1182/blood-2002-05-1320
25. Jurrmann N, Brigelius-Flohe R, Bol GF (2005) Curcumin blocks
interleukin-1 (IL-1) signaling by inhibiting the recruitment of the
IL-1 receptor-associated kinase IRAK in murine thymoma EL-4
cells. J Nutr 135(8):18591864
26. Biswas SK,McClure D, Jimenez LA, Megson IL, Rahman I (2005)
Curcumin induces glutathione biosynthesis and inhibits NF-kappa
B activation and interleukin-8 release in alveolar epithelial cells:
mechanism of free radical scavenging activity. Antioxid Redox
Sign 7(12):3241. doi:10.1089/ars.2005.7.32
27. Leu TH, Su SL, Chuang YC, Maa MC (2003) Direct inhibitory
effect of curcumin on src and focal adhesion kinase activity.
Biochem Pharmacol 66(12):23232331
28. Chen YR, Tan TH (1998) Inhibition of the c-Jun N-terminal kinase
(JNK) signaling pathway by curcumin. Oncogene 17(2):173178.
doi:10.1038/sj.onc.1201941
29. Vietri M, Pietrabissa A, Mosca F, Spisni R, Pacifici GM (2003)
Curcumin is a potent inhibitor of phenol sulfotransferase
(SULT1A1) in human liver and extrahepatic tissues. Xenobiotica
33(4):357363. doi:10.1080/0049825031000065197
30. Reddy S, Aggarwal BB (1994) Curcumin is a noncompetitive and
selective inhibitor of phosphorylase-kinase. FEBS Lett 341(1):19
22. doi:10.1016/0014-5793(94)80232-7
31. Fu XY, Yang MF, Cao MZ, Li DW, Yang XY, Sun JY, Zhang ZY,
Mao LL et al (2016) Strategy to suppress oxidative damage-
induced neurotoxicity in PC12 cells by curcumin: the role of
ROS-mediated DNA damage and the MAPK and AKT pathways.
Mol Neurobiol 53(1):369378. doi:10.1007/s12035-014-9021-1
32. Dai CS, Li JC, Li J (2013) New insight in colistin induced neuro-
toxicity with the mitochondrial dysfunction in mice central nervous
tissues. Exp Toxicol Pathol 65(6):941948. doi:10.1016/j.
etp.2013.01.008
33. Hara H, Ohta M, Adachi T (2006) Apomorphine protects against 6-
hydroxydopamine-induced neuronal cell death through activation
of the Nrf2-ARE pathway. J Neurosci Res 84(4):860866.
doi:10.1002/jnr.20974
34. WangL,JiangHY,YinZB,AschnerM,CaiJY(2009)
Methylmercury toxicity and Nrf2-dependent detoxification in as-
trocytes. Toxicol Sci 107(1):135143. doi:10.1093/toxsci/kfn201
Mol Neurobiol
35. Allam R, Darisipudi MN, Rupanagudi KV, Lichtnekert J, Tschopp
J, Anders HJ (2011) Cutting edge: cyclic polypeptide and amino-
glycoside antibiotics trigger IL-1 beta secretion by activating the
NLRP3 Inflammasome. J Immunol 186(5):27142718.
doi:10.4049/jimmunol.1002657
36. Jaber BL, Sundaram S, Neto MC, King AJ, Pereira BJG (1998)
Polymyxin-B stimulates tumor necrosis factor-alpha production
by human peripheral blood mononuclear cells. Int J Artif Organs
21(5):269273
37. Karunaweera N, Raju R, Gyengesi E, Munch G (2015) Plant poly-
phenols as inhibitors of NF-kappa B induced cytokine production a
potential anti-inflammatory treatment for Alzheimers disease?
Front Mol Neurosci 8. doi: 10.3389/fnmol.2015.00024
38. Sung B, Pandey MK, Ahn KS, Yi T, Chaturvedi MM, Liu M,
Aggarwal BB (2008) Anacardic acid (6-nonadecyl salicylic acid),
an inhibitor of histone acetyltransferase, suppresses expression of
nuclear factor-kappaB-regulated gene products involved in cell sur-
vival, proliferation, invasion, and inflammation through inhibition
of the inhibitory subunit of nuclear factor-kappaBalpha kinase,
leading to potentiation of apoptosis. Blood 111(10):48804891.
doi:10.1182/blood-2007-10-117994
39. Cheng AL,Hsu CH, Lin JK, Hsu MM, Ho YF, Shen TS, Ko JY, Lin
JT et al (2001) Phase I clinical trial of curcumin, a chemopreventive
agent, in patients with high-risk or pre-malignant lesions.
Anticancer Res 21(4B):28952900
40. Sharma RA, McLelland HR, Hill KA, Ireson CR, Euden SA,
Manson MM, Pirmohamed M, Marnett LJ et al (2001)
Pharmacodynamic and pharmacokinetic study of oral curcuma
extract in patients with colorectal cancer. Clin Cancer Res 7(7):
18941900
41. Kumar A, Takada Y, Boriek AM, Aggarwal BB (2004) Nuclear
factor-kappaB: its role in health and disease. J Mol Med (Berl)
82(7):434448. doi:10.1007/s00109-004-0555-y
42. Jobin C, Bradham CA, Russo MP, Juma B, Narula AS, Brenner
DA, Sartor RB (1999) Curcumin blocks cytokine-mediated NF-
kappa B activation and proinflammatory gene expression by
inhibiting inhibitory factor I-kappa B kinase activity. J Immunol
163(6):34743483
43. Yamashita Y, Ueyama T, Nishi T, Yamamoto Y, Kawakoshi A,
Sunami S, Iguchi M, Tamai H et al (2014) Nrf2-inducing anti-ox-
idation stress response in the rat livernew beneficial effect of
lansoprazole. PLoS One 9(5):e97419. doi:10.1371/journal.
pone.0097419
44. Karunaweera N, Raju R, Gyengesi E, Munch G (2015) Plant poly-
phenols as inhibitors of NF-kappaB induced cytokine production-a
potential anti-inflammatory treatment for Alzheimers disease?
Front Mol Neurosci 8:24. doi:10.3389/fnmol.2015.00024
45. Plummer SM, Holloway KA, Manson MM, Munks RJ, Kaptein A,
Farrow S, Howells L (1999) Inhibition of cyclo-oxygenase 2 ex-
pression in colon cells by the chemopreventive agent curcumin
involves inhibition of NF-kappaB activation via the NIK/IKK sig-
nalling complex. Oncogene 18(44):60136020. doi:10.1038/sj.
onc.1202980
46. Shishodia S, Aggarwal BB (2002) Nuclear factor-kappaB activa-
tion: a question of life or death. J Biochem Mol Biol 35(1):2840
47. Pahl HL (1999) Activators and target genes of Rel/NF-
kappaB transcription factors. Oncogene 18(49):68536866.
doi:10.1038/sj.onc.1203239
48. Bellezza I, Tucci A, Galli F, Grottelli S, Mierla AL, Pilolli F,
Minelli A (2012) Inhibition of NF-kappa B nuclear transloca-
tion via HO-1 activation underlies alpha-tocopheryl succinate
toxicity. J Nutr Biochem 23(12):15831591. doi:10.1016/j.
jnutbio.2011.10.012
49. Gou M, Men K, Shi H, Xiang M, Zhang J, Song J, Long J, Wan Y
et al (2011) Curcumin-loaded biodegradable polymeric micelles for
colon cancer therapy in vitro and in vivo. Nanoscale 3(4):1558
1567. doi:10.1039/c0nr00758g
50. Aggarwal BB, Surh Y-J, Shishodia S (2007) The molecular targets
and therapeutic uses of curcumin in health and disease, vol 595.
Springer, New York
51. Gupta SC, Patchva S, Aggarwal BB (2013) Therapeutic roles of
curcumin: lessons learned from clinical trials. Aaps Journal 15(1):
195218. doi:10.1208/s12248-012-9432-8
Mol Neurobiol
... Typically, these symptoms decrease upon tapering or discontinuation of the drug. The administration of colistin triggers the activation of pro-inflammatory mediators within neuronal cells [145]. Research indicates that neurotoxicity entails a complex interplay of apoptotic and inflammatory pathways. ...
... Studies involving colistin treatment (15 mg/kg/day for 7 days) revealed significant mitochondrial dysfunction in central and peripheral nervous tissues [146,147]. Similarly, exposure to colistin (200 μM/24 h) induced apoptosis in around 50% of neuronal N2a cells in mice [145]. Further exploration using Western blotting and immunohistochemistry demonstrated that colistin-induced apoptosis in N2a neuronal cells hinges on generating reactive oxygen species (ROS) and the mitochondrial pathway [145,148,149]. ...
... Similarly, exposure to colistin (200 μM/24 h) induced apoptosis in around 50% of neuronal N2a cells in mice [145]. Further exploration using Western blotting and immunohistochemistry demonstrated that colistin-induced apoptosis in N2a neuronal cells hinges on generating reactive oxygen species (ROS) and the mitochondrial pathway [145,148,149]. Interestingly, co-administration of neuroprotective agents, such as curcumin and minocycline demonstrated, in vivo efficacy against polymyxininduced neurotoxicity [145,149]. ...
Chapter
Full-text available
Nosocomial infections caused by carbapenem-resistant Acinetobacter baumannii (CRAB) have become a global concern. The extensive antibiotic resistance of CRAB has significantly limited treatment options, while its prevalence in hospital outbreaks has amplified infection rates. This scenario has led to a resurgence of interest in polymyxins, an older class of antibiotics previously overlooked due to perceived toxicity. Polymyxins, cationic polypeptide antibiotics, now represent a last-resort treatment option. Despite their historical use, modern assessment methods have only recently been applied to evaluate polymyxins. Two polymyxins are available for clinical use: polymyxin B and colistin (polymyxin E). Notably, the administration of these drugs is hindered by toxicities, primarily nephrotoxicity and neurotoxicity, alongside less common adverse effects such as injection pain, hypersensitivity reactions, and bronchospasms.
... Recent studies indicate that colistin-induced neurotoxicity is mainly related to mitochondrial dysfunction and oxidative stress (Dai et al. 2017b, Dai et al. 2013a, Liu et al. 2013, Dai et al. 2018b, Dai et al. 2016, Dai et al. 2013b), although the data shows that multiple inflammatory and pro/anti-apoptotic signaling pathways including PI3K, Akt, p53, mitogenactivated protein kinase, adenosine monophosphate (AMP)activated protein kinase, mammalian target of rapamycin C1 (mTORC1), NRF2/HO-11, Bax, and Bcl-2 are involved in colistin-induced neuronal cell death as well (Dai et al. 2017b, Dai et al. 2016, Dai et al. 2013b, Dai et al. 2018a, Dai et al. 2017a). The consequences of colistin neurotoxicity on different markers are shown in Fig. 2. ...
... It involves the downregulation of NF-kB signaling and the mediators of inflammation (e.g., COX-2) by inhibiting the activity of IKK kinase. So, this investigation supports the neuroprotective effects of curcumin during colistin treatment (Dai et al. 2018a;Dai et al. 2020b). ...
Article
Full-text available
Polymyxin E or colistin is an effective antibiotic against MDR Gram-negative bacteria. Due to unwanted side effects, the use of this antibiotic has been limited for a long time, but in recent years, the widespread of MDR Gram-negative bacteria infections has led to its reintroduction. Neurotoxicity and nephrotoxicity are the significant dose-limiting adverse effects of colistin. Several agents with anti-inflammatory and antioxidant properties have been used for the prevention of colistin-induced neurotoxicity. This study aims to review the preclinical studies in this field to prepare guidance for future human studies. The data was achieved by searching PubMed, Scopus, and Google Scholar databases. All eligible pre-clinical studies performed on neuroprotective agents against colistin-induced neurotoxicity, which were published up to September 2023, were included. Finally, 16 studies (ten in vitro and eight in vivo) are reviewed. Apoptosis (in 13 studies), inflammatory (in four studies), and oxidative stress (in 14 studies) pathways are the most commonly reported pathways involved in colistin-induced neurotoxicity. The assessed compounds include non-herbal (e.g., ascorbic acid, rapamycin, and minocycline) and herbal (e.g., curcumin, rutin, baicalein, salidroside, and ginsenoside) agents. Besides these compounds, some other measures like transplantation of mitochondria and the use of nerve growth factor and mesenchymal stem cells could be motivating subjects for future research. Based on the data from experimental (in vitro and animal) studies, a combination of colistin with neuroprotective agents could prevent or decrease colistin-induced neurotoxicity. However, well-designed randomized clinical trials and human studies are essential for demonstrating efficacy.
... Combination of curcumin with (-)-epigallocatechin gallate (EGCG), the most active component of tea, was more efficient in inhibiting biofilm formation by a number of wastewater bacteria [79]. In addition to direct bactericidal/static effects, mechanisms of action include cell membrane disruption, interference with quorum sensing, inhibition of biofilm formation, inhibition of cell division, induction of oxidative stress, induction of programmed cell death, phototoxicity, modulation of bacterial cell metabolism, and inhibition of the intracellular proliferation of bacteria [35,75,80]. The antimicrobial action of curcumin against Salmonella also involves reducing the number of flagella by directly binding to them and making them unstable [81]. ...
Article
Full-text available
Curcumin, a natural polyphenolic component from Curcuma longa roots, is the main bioactive component of turmeric spice and has gained increasing interest due to its proposed anti-cancer, anti-obesity, anti-inflammatory, antioxidant, and lipid-lowering effects, in addition to its thermogenic capacity. While intake from dietary sources such as curry may be sufficient to affect the intestinal microbiome and thus may act indirectly, intact curcumin in the body may be too low (<1 microM) and not sufficient to affect signaling and gene expression, as observed in vitro with cultured cells (10–20 microM). Several strategies can be envisioned to increase curcumin levels in the body, such as decreasing its metabolism or increasing absorption through the formation of nanoparticles. However, since high curcumin levels could also lead to undesired regulatory effects on cellular signaling and gene expression, such studies may need to be carefully monitored. Here, we review the bioavailability of curcumin and to what extent increasing curcumin levels using nanoformulations may increase the bioavailability and bioactivity of curcumin and its metabolites. This enhancement could potentially amplify the disease-preventing effects of curcumin, often by leveraging its robust antioxidant properties.
... Curcuminoids have gained potential therapeutic interest for the treatment of immune-related metabolic disorders and cancer due to their wide spectrum of biological targets and absence of adverse effects. [6][7][8][9][10][11][12][13][14][15]. We looked at the antibacterial, anticancer, and antioxidant characteristics of thaizolyl dispersion dyes. ...
... In this study, the cytotoxic effects of AFB1 and T-2 on PK15 cells were investigated using different methods to determine if oxidative stress played a role. Oxidative stress, induced by the generation of ROS, is a major mechanism of neurotoxicity for several envi-ronmental and food contaminants, including the T-2 toxin, deoxynivalenol, and fumonisin B [35][36][37][38][39][40][41]. This process causes damage to DNA, promotes lipid peroxidation, protein damage, and ultimately leads to cell death. ...
Article
Full-text available
In China, animal feeds are frequently contaminated with a range of mycotoxins, with Aflatoxin B1 (AFB1) and T-2 toxin (T-2) being two highly toxic mycotoxins. This study investigates the combined nephrotoxicity of AFB1 and T-2 on PK15 cells and murine renal tissues and their related oxidative stress mechanisms. PK15 cells were treated with the respective toxin concentrations for 24 h, and oxidative stress-related indicators were assessed. The results showed that the combination of AFB1 and T-2 led to more severe cellular damage and oxidative stress compared to exposure to the individual toxins (p < 0.05). In the in vivo study, pathological examination revealed that the kidney tissue of mice exposed to the combined toxins showed signs of glomerular atrophy. The contents of oxidative stress-related indicators were significantly increased in the kidney tissue (p < 0.05). These findings suggest that the combined toxins cause significant oxidative damage to mouse kidneys. The study highlights the importance of considering the combined effects of mycotoxins in animal feed, particularly AFB1 and T-2, which can lead to severe nephrotoxicity and oxidative stress in PK15 cells and mouse kidneys. The findings have important implications for animal feed safety and regulatory policy.
Article
Full-text available
Aflatoxin (AFT) contamination poses a significant global public health and safety concern, prompting widespread apprehension. Of the various AFTs, aflatoxin B1 (AFB1) stands out for its pronounced toxicity and its association with a spectrum of chronic ailments, including cardiovascular disease, neurodegenerative disorders, and cancer. Lycopene, a lipid-soluble natural carotenoid, has emerged as a potential mitigator of the deleterious effects induced by AFB1 exposure, spanning cardiac injury, hepatotoxicity, nephrotoxicity, intestinal damage, and reproductive impairment. This protective mechanism operates by reducing oxidative stress, inflammation, and lipid peroxidation, and activating the mitochondrial apoptotic pathway, facilitating the activation of mitochondrial biogenesis, the endogenous antioxidant system, and the nuclear factor erythroid 2-related factor 2 (Nrf2)/kelch-like ECH-associated protein 1 (KEAP1) and peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) pathways, as well as regulating the activities of cytochrome P450 (CYP450) enzymes. This review provides an overview of the protective effects of lycopene against AFB1 exposure-induced toxicity and the underlying molecular mechanisms. Furthermore, it explores the safety profile and potential clinical applications of lycopene. The present review underscores lycopene’s potential as a promising detoxification agent against AFB1 exposure, with the intent to stimulate further research and practical utilization in this domain.
Article
Full-text available
Antibiotic colistin is the last line of defense against multidrug‐resistant (MDR) Gram‐negative bacterial infections. Emergence of colistin resistance in microbes is a critical challenge. Herein, curcumin is discovered, for the first time, to reverse the resistance phenotype of colistin‐resistant bacteria via a checkerboard assay. For the co‐delivery of curcumin and colistin, negatively charged poly(ethylene glycol)‐functionalized liposomes encapsulating both drugs (Lipo‐cc) are prepared. Killing kinetics and live/dead assays confirm the antibacterial activity of Lipo‐cc against colistin‐resistant bacteria, which is more potent than that of the free curcumin and colistin combination. Mechanistical studies reveal that Lipo‐cc restores the affinity of colistin for the bacterial membrane and improves the uptake of curcumin, which leads to reduced efflux pump activity, achieving a synergistic effect of colistin and curcumin. At the effective antibacterial dose, Lipo‐cc does not exhibit any toxicity. The therapeutic efficacy of Lipo‐cc is further demonstrated in an intestinal bacterial infection model induced with colistin‐resistant Escherichia coli. Lipo‐cc reduces the bacterial burden with over 6‐log reduction and alleviated inflammation caused by infection. Importantly, unlike colistin, Lipo‐cc does not affect the homeostasis of the intestinal flora. Taken together, Lipo‐cc successfully overcame colistin resistance, indicating its potential for the treatment of colistin‐resistant bacterial infections.
Article
Full-text available
Polymyxins B and E (i.e., colistin) are a family of naturally occurring lipopeptide antibiotics that are our last line of defense against multidrug resistant (MDR) Gramnegative pathogens. Unfortunately, nephrotoxicity is a dose limiting factor for polymyxins that limits their clinical utility. Our recent studies demonstrate that polymyxin-induced nephrotoxicity is a result of their extensive accumulation in renal tubular cells. The design and development of safer, novel polymyxin lipopeptides is hampered by our limited understanding of their complex structure nephrotoxicity relationships. This is the first study to employ a novel targeted chemical biology approach to map the polymyxin recognition epitope of a commercially available polymyxin mAb and demonstrate its utility for mapping the kidney distribution of a novel, less nephrotoxic polymyxin lipopeptide. Eighteen novel polymyxin lipopeptide analogues were synthesized with modifications in the polymyxin core domains, namely, the N-terminal fatty acyl region, tripeptide linear segment, and cyclic heptapeptide. Surface plasmon resonance epitope mapping revealed that the monoclonal antibody (mAb) recognition epitope consisted of the hydrophobic domain (N-terminal fatty acyl and position 6/7) and diaminobutyric acid (Dab) residues at positions 3, 5, 8, and 9 of the polymyxin molecule. Structural diversity within the hydrophobic domains and Dab 3 position are tolerated. Enlightened with an understating of the structure binding relationships between the polymyxin mAb and the core polymyxin scaffold, we can now rationally employ the mAb to probe the kidney distribution of novel polymyxin lipopeptides. This information will be vital in the design of novel, safer polymyxins through chemical tailoring of the core scaffold and exploration of the elusive/complex polymyxin structure nephrotoxicity relationships.
Article
Full-text available
Neurotoxicity remains a poorly characterized adverse effect associated with colistin therapy. The aim of the present study was to investigate the mechanism of colistin-induced neurotoxicity using the mouse neuroblastoma2a (N2a) cell line. Colistin treatment (0-200 μM) of N2a neuronal cells induced apoptotic cell death in a dose-dependent manner. Colistin-induced neurotoxicity was associated with a significant increase of reactive oxygen species (ROS) levels, with a concomitant decrease in the activities of superoxide dismutase (SOD), catalase (CAT), and the glutathione (GSH) levels. Mitochondrial dysfunction was evident from the dissipation of membrane potential and the increase of Bax/Bcl-2, followed by the release of cytochrome c (CytC). Caspase-3/7, -8, and -9 activations were also detected. Colistin-induced neurotoxicity significantly increased the gene expression of p53 (1.6-fold), Bax (3.3-fold), and caspase-8 (2.2-fold) (all p < 0.01). The formation of autophagic vacuoles was evident with the significant increases (all p < 0.05 or 0.01) of both of Beclin 1 and LC3B following colistin treatment (50-200 μM). Furthermore, inhibition of autophagy by pretreatment with chloroquine diphosphate (CQ) enhanced colistin-induced apoptosis via caspase activation, which could be attenuated by co-treatment with the pan-caspase inhibitor Z-VAD-FMK. In summary, our study reveals that colistin-induced neuronal cell death involves ROS-mediated oxidative stress and mitochondrial dysfunction, followed by caspase-dependent apoptosis and autophagy. A knowledge base of the neuronal signaling pathways involved in colistin-induced neurotoxicity will greatly facilitate the discovery of neuroprotective agents for use in combination with colistin to prevent this undesirable side effect.
Article
Full-text available
Alzheimer's disease (AD) is a neurodegenerative disorder that impacts the daily lives of many sufferers through memory loss as well as behavioral and cognitive changes. AD is the most common form of dementia. One in ten people over the age of 65, and around half of those over 85 have AD. AD can be divided into familial (early-onset) and sporadic (late-onset) cases, with the familial form (<1%) linked to mutations in three major genes (amyloid precursor protein, presenilin-1 and 2), and the sporadic form (>99% of cases) caused by a variety of genetic (e.g., apolipoprotein E), metabolic and environmental factors. The AD brain is characterized macroscopically by cortical atrophy, caused by degeneration of the cholinergic axonal arborisation and shrinkage of the dendritic tree. Microscopically, amyloid beta peptide deposits (senile plaques) and neurofibrillary tangles are present in affected areas (Gil-Bea et al., 2012). AD is also characterized by chronic neuroinflammation, driven by activation of astroglia and microglia (Rosenblum, 2014). In addition, levels of pro-inflammatory mediators or cytokines which include chemokines, interferons, interleukins, lymphokines, and tumor necrosis factors are elevated in the brains of patients with AD (Latta et al., 2014). Furthermore, nuclear translocation of NF-κB and STAT-1α, transcription factors involved in pro-inflammatory gene expression, indicates the presence of a sustained pro-inflammatory process (Lawrence, 2009). Drug Discovery Difficulties Faced in Alzheimer's Disease Drug discovery for AD has been strongly focused on β-amyloid (initially plaques, then soluble oligomers), as genetic evidence from the familial cases supported by the hypothesis that β-amyloid must be driving the disease process. Based on the " amyloid cascade hypothesis, " anti-amyloid therapies were hoped to deliver a cure for AD (Robinson et al., 2004). Unfortunately, numerous clinical trials with active and passive amyloid vaccines as well as G-secretase inhibitors have failed (reviewed in Castello et al., 2014). Currently, there are no disease-modifying drugs available for AD. Consequently, alternative therapeutic targets, such as neuroinflammation have been suggested for the prevention and treatment of AD (Shi et al., 2013; Latta et al., 2014). As the expression of many pro-inflammatory cytokines is driven by the transcription factor NF-κB (Hoffmann et al., 2006), we propose that brain-permeable inhibitors of NF-κB signaling have the potential to prevent or slow down the progression of AD.
Book
Curcumin is derived from the root of the plant Curcuma longa (also called turmeric) and its medicinal uses have been described for over 5000 years. More than 1500 papers published within last half a century has revealed that curcumin has a potential in the treatment of wide variety of inflammatory diseases including cancer, diabetes, cardiovascular diseases, arthritis, Alzheimer, psoriasis etc, through modulation of numerous molecular targets. This is the first monograph to deal specifically with this subject.
Article
Scientific research provides documented evidence that fatty acid metabolites have profound impact on carcinogenesis. Intervention into dioxygenase pathways might therefore effect development, metastasis and progression of man types of cancers. This work delivers the first 3D structural data and explains how curcumin interacts with the fatty acid metabolizing enzyme, soybean lipoxygenase. Curcumin binds to lipoxygenase in a noncompetitive manner. Trapped in that complex, it undergoes photodegradation in the X-rays, but utilizes enzyme catalytic ability to form the peroxy complex Enz-Fe-O-O-R as 4-hydroperoxy-2-methoxy-phenol, that later transforms into 2-methoxycyclohexa-2,5-diene-1,4-dione. Our observations about this radiation and time-dependent inhibition add new information to the role that curcumin might play in cancer prevention and treatment.
Article
Amyloid beta (Aβ) peptide is the major constituent of the extracellular amyloid plaques deposited in the brains of Alzheimer's disease patients and is central to the pathogenic pathway causing this disease. The identity of the neurotoxic Aβ species remains elusive. We previously reported that Aβ toxicity correlates strongly with its neuronal cell binding leading us to hypothesize that neuronal cell death is caused by the binding of a specific oligomeric Aβ species. To identify the specific oligomeric Aβ species that is associated with cell death, we treated mouse cortical neuronal cultures with synthetic Aβ40 and Aβ42 peptides and identified that the cellular Aβ binding and neurotoxicity were time and concentration dependent. We found a significant correlation between the amount of trimer and tetramer species bound to neurons with increasing neurotoxicity. We prepared Aβ40 oligomers (up to tetramers) using photo‐induced cross‐linking of unmodified peptides to confirm this oligomer‐specific neurotoxic activity. Our results identify the Aβ tetramer, followed by the trimer, as the most toxic low‐order oligomers Aβ species. image Our findings suggested that binding of amyloid‐β (Aβ) tetramer and trimer, not monomer or dimer, to neurons is critical to induce neuronal cell death associated with Alzheimer's Disease. We proposed that Aβ trimer and tetramer are the potential neurotoxic Aβ species. This would provide more specific therapeutic target for Alzheimer's Disease . Read the Editorial Highlight for this article on page 437 .
Article
The concept of using phytochemicals has ushered in a new revolution in pharmaceuticals. Naturally occurring polyphenols (like curcumin, morin, resveratrol, etc.) have gained importance because of their minimal side effects, low cost and abundance. Curcumin (diferuloylmethane) is a component of turmeric isolated from the rhizome of Curcuma longa. Research for more than two decades has revealed the pleiotropic nature of the biological effects of this molecule. More than 7000 published articles have shed light on the various aspects of curcumin including its antioxidant, hypoglycemic, anti-inflammatory and anti-cancer activities. Apart from these well-known activities, this natural polyphenolic compound also exerts its beneficial effects by modulating different signalling molecules including transcription factors, chemokines, cytokines, tumour suppressor genes, adhesion molecules, microRNAs, etc. Oxidative stress and inflammation play a pivotal role in various diseases like diabetes, cancer, arthritis, Alzheimer's disease and cardiovascular diseases. Curcumin, therefore, could be a therapeutic option for the treatment of these diseases, provided limitations in its oral bioavailability can be overcome. The current review provides an updated overview of the metabolism and mechanism of action of curcumin in various organ pathophysiologies. The review also discusses the potential for multifunctional therapeutic application of curcumin and its recent progress in clinical biology. Copyright © 2015. Published by Elsevier Ltd.
Article
We aimed to determine the effects of curcumin on liver fibrosis and to clarify the role of nuclear factor-κB (NF-κB) and inducible nitric oxide synthase (iNOS) in the model of microsurgical cholestasis in early stage of extrahepatal biliary atresia. 12-week-old Wistar rats were divided into four groups (n=8): sham operated rats (received olive oil after laparotomy); curcumin group (received curcumin, 200mg/kg/day, after laparotomy); biliary duct ligated group (BDL, received olive oil after operation); biliary duct ligated group (BDL curc, received curcumin, 200mg/kg/day, after operation). After 3 weeks of the treatment, curcumin did not modify blood plasma markers as well as iNOS and NF-κB (p65) expressions in the liver species of the sham group. Interestingly, there was significant increase in both liver and kidney fibrosis extents. On the other hand, despite decrease in iNOS and NF-κB (p65) expressions, curcumin treatment did not affect fibrosis enlargement due to bile duct ligation in the liver. In the BDL group, curcumin treatment decreased the level of blood plasma markers investigated. In conclusion, curcumin treatment was able to improve functional properties of hepatocytes and to inhibit both NF-κB and iNOS upregulations in the BDL group, yet without beneficial effect on the liver fibrosis developed in this model of cholestasis. Thus, in the studied model of microsurgical cholestasis, other factors, different from NF-κB and iNOS are responsible for fibrotic process in the liver.
Article
In the face of diminishing therapeutic options for the treatment of infections caused by multidrug-resistant, Gram-negative bacteria, dinicians are increasingly using colistin and polymyxin B. These antibiotics became available clinically in the 1950s, when understanding of antimicrobial pharmacology and regulatory requirements for approval of drugs was substantially less than today. At the 1st International Conference on Polymyxins in Prato, Italy, 2013, participants discussed a set of key objectives that were developed to explore the factors affecting the safe and effective use of polymuxins, identify the gaps in knowledge, and set priorities for future research. Participants identified several factors that affect the optimum use of polymyxins, including: confusion caused by several different conventions used to describe doses of colistin; an absence of appropriate pharmacopoeial standards for polymyxins; outdated and diverse product information; and uncertainties about susceptibility testing and breakpoints. High-priority areas for research included: better definition of the effectiveness of polymyxin-based combination therapy compared with monotherapy via well designed, randomised controlled trials; examination of the relative merits of colistin versus polymyxin B for various types of infection; investigation of pharmacokinetics in special patient populations; and definition of the role of nebulised polymyxins alone or in combination with intravenous polymyxins for the treatment of pneumonia. The key areas identified provide a roadmap for action regarding the continued use of polymyxins, and are intended to help with the effective and safe use of these important, last-line antibiotics.
Article
Oxidative damage plays a key role in causation and progression of neurodegenerative diseases. Inhibition of oxidative stress represents one of the most effective ways in treating human neurologic diseases. Herein, we evaluated the protective effect of curcumin on PC12 cells against H2O2-induced neurotoxicity and investigated its underlying mechanism. The results indicated that curcumin pre-treatment significantly suppressed H2O2-induced cytotoxicity, inhibited the loss of mitochondrial membrane potential (Δψm) through regulation of Bcl-2 family expression, and ultimately reversed H2O2-induced apoptotic cell death in PC12 cells. Attenuation of caspase activation, poly(ADP-ribose) polymerase (PARP) cleavage, DNA damage, and accumulation of reactive oxygen species (ROS) all confirmed its protective effects. Moreover, curcumin markedly alleviated the dysregulation of the MAPK and AKT pathways induced by H2O2. Taken together, our findings suggest that the strategy of using curcumin could be a highly effective way in combating oxidative damage-mediated human neurodegenerative diseases.