Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2017

Open Access 01.12.2017 | Research article

Postnatal treatment using curcumin supplements to amend the damage in VPA-induced rodent models of autism

verfasst von: Maha Al-Askar, Ramesa Shafi Bhat, Manar Selim, Laila Al-Ayadhi, Afaf El-Ansary

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2017

Abstract

Background

Valproic acid (VPA) is used as a first-line antiepileptic agent and is undergoing clinical trials for use as a treatment for many disorders. Mothers undergoing VPA treatment during early pregnancy reportedly show increased rates of autism among their offspring. The benefits of curcumin supplementation were investigated using an animal model of VPA-induced autism.

Methods

The study was performed using a rodent model of autism by exposing rat fetuses to valproic acid (VPA) on the 12.5th day of gestation. At 7 days from their birth, the animals were supplemented with a specific dose of curcumin. Forty neonatal male Western Albino rats were divided into four groups. Rats in group I received only phosphate-buffered saline, rats in group II were the prenatal VPA exposure newborns, rats in group III underwent prenatal VPA exposure supplemented with postnatal curcumin, and rats in group IV were given only postnatal curcumin supplements.

Results

VPA rats exhibited delayed maturation and lower body and brain weights with numerous signs of brain toxicity, such as depletion of IFN-γ, serotonin, glutamine, reduced glutathione, glutathione S-transferase, lipid peroxidase with an increase in CYP450, IL-6, glutamate, and oxidized glutathione. A curcumin supplement moderately corrected these dysfunctions and was especially noticeable in improving delayed maturation and abnormal weight.

Conclusions

Curcumin plays a significant therapeutic role in attenuating brain damage that has been induced by prenatal VPA exposure in rats; however, its therapeutic role as a dietary supplement still must be certified for use in humans.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12906-017-1763-7) contains supplementary material, which is available to authorized users.

Background

Experimental animal models of autism can help researchers understand the etiology of autism in humans and explore various supplements used to amend the impaired biomarkers related to the disease [1]. In reality, autism manifests as a set of behavioral changes, and the behavior of an animal model can never be the same as the behavior of an autistic child, but these behaviors can be scrutinized using precise experiments to measure the behavioral modifications. [2, 3] Currently, different approaches are used to induce human-like autistic features in rodent models by exposing animals to certain chemicals, such as valproic acid (VPA), since VPA significantly increases the rate of autism among the offspring of human mothers who are medicated with VPA during early pregnancy [4]. VPA exposure during the first trimester of conception is associated with risk of autism in the child, particularly if exposure occurs during the time of neural tube closure [5]. Thus, our rodent models showing autistic features were male Wistar neonatal rats originating from valproate-treated females [6]. These females received a single intraperitoneal injection of 600 mg/kg of sodium valproate on day 12.5 after conception.
For many years, VPA, a branched short-chain fatty acid, was used as a first-line antiepileptic agent, particularly in children suffering from epilepsy. [7] Presently, this drug is in clinical trials for use in the treatment of many disorders, however various consequences such as fatal hemorrhaging, pancreatitis, bone marrow suppression, hepatotoxicity, and hyperammonemic encephalopathy are associated with its use. VPA acts on γ amino butyric acid (GABA) levels, changes the activity of many neurotransmitters, and blocks Na + channels, Ca2+ channels and voltage-gated channels in brain tissue [8]. Many studies have shown that valproate exposure in utero is associated with increased risk of neural tube defects, neurodevelopmental deficits and reduced vocal skills. [911].
Curcumin is known for its protective actions against various central nervous system disorders such as Alzheimer’s disease, tardive dyskinesia, major depression, epilepsy, neurodegenerative disorders and neuropsychiatric disorders [12]. It can cross the blood brain barrier and is nontoxic at high doses [13]. Many studies have proved that curcumin targets multiple degenerative pathways including oxidative/nitrosative stress, mitochondrial dysfunction, and protein aggregation [14]. Curcumin was effective in ameliorating propionic acid-induced autism in rats through the suppression of oxidative stress, mitochondrial dysfunction and neuroinflammation [14]. All reported biological activities of curcumin could potentially be of interest as autism therapies. Therefore, we studied the therapeutic effects of curcumin in VPA-induced animal models of autism.

Methods

Chemicals

Curcumin from Curcuma longa (Turmeric) (powder, 50 g in a glass bottle) and valproic acid sodium salt (powder, 25 g in a glass bottle) were obtained from Sigma. The catalog numbers are C1386 and P4543, respectively.

Animals

Female Wistar rats (180–200 g) that were acclimated in our laboratory under standard laboratory conditions with a controlled fertility cycle were obtained from the Center for Laboratory Animals and Experimental Surgery at King Khalid University Hospital, Riyadh. Rats were mated overnight, and the pregnant rats were divided into 2 sets. On day 12.5 after conception, set I was injected with a single intraperitoneal injection of normal saline, and set II was injected with a single intraperitoneal injection of 600 mg/kg of sodium valproate [15]. Twenty Male Wistar neonatal rats that were born from each set of females (treated with normal saline and VPA) were further divided into two groups of ten pups each. Finally, four groups (10 neonatal rats each) were organized as follows:
Group I (control): Male pups from set I was given an oral dose of 1 ml of normal saline on day 7 after birth.
Group II (VPA rodent model): Male pups from set II (valproate treated mothers) were given an oral dose of 1 ml of normal saline on day 7 after birth.
Group III (VPA-Curcumin): Male pups from set II (valproate treated mothers) received 1 ml of curcumin (1 g/kg b.wt) [16] orally at 7 days after birth.
Group IV (Curcumin): Male pups from set I received 1 ml of curcumin (1 g/kg b.wt) [16] orally at 7 days after birth. Figure 1 summarizes the experimental design and the different groups that were studied.

Postnatal growth and maturation development

Animals were weighed at 0, 7, 14, 21 and 27 days after delivery. See the uploaded diagram that illustrates the experimental design.

Tissue preparation

On the 28th day after birth, all groups were killed by decapitation. The brains were quickly collected, weighed, washed with normal saline and then homogenized in 10 times w/v bi-distilled water and further used in the biochemical assays.

Biochemical analyses

Tissue samples were run concordant with the instructions of the kit protocol. The quantification of interleukin-6, interferon gamma, and reduced glutathione in the brain tissue were determined using a rat ELISA Kit obtained from “My Bio Source” that were based on a quantitative sandwich immunoassay technique, while for cytochrome P450, enzyme-linked immune sorbent assay, based on the biotin double antibody sandwich technology obtained from “My Bio Source”, was used. The quantification of lipid peroxide, glutathione S-transferases, glutamine, and glutamate in the brain tissue was determined using the ELISA Kit based on a quantitative sandwich immunoassay technique obtained from “Cusabioin”.
The quantification of serotonin in the brain tissue was carried out using a 5-HT ELISA Kit, which applies the competitive enzyme immunoassay technique utilizing a monoclonal anti-5-HT antibody and a 5HT-HRP conjugate, and for oxidized glutathione, a GSSG ELISA kit was used, which applies the competitive enzyme immunoassay technique utilizing a monoclonal anti-GSSG antibody and a GSSG-HRP conjugate, which were obtained from My Bio Source.

Statistical analysis

The Statistical Package for the Social Sciences (SPSS) computer program was used. The results were expressed as the mean ± S.D., and all statistical comparisons were made using independent T-Tests, with values of P ≤ 0.05 considered to be significant. Pearson’s correlations were also performed, and the best fit line was drawn. Receiver operating characteristics (ROC) analysis was performed. Area under the curve, cutoff values threshold, and degrees of specificity and sensitivity were calculated.

Results

The analysis of the body weight, brain weight and eye opening age in pups showed statistically significant (P < 0.001) differences in all tested groups compared with the control group, as shown in Table 1. VPA-exposed rats showed delayed maturation, as represented by lower body weight, a slight decrease in brain weight and late eye opening compared to the control group, whereas curcumin treatment was effective in promoting body and brain weight in VPA-exposed pups (Table 1).
Table 1
Mean ± S.D. together with the independent t-test for weight gain, brain weight and age of eye opening between neurointoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameter
Group
N
Min
Max
Mean ± S.D.
Percent Change
P valuea
P valueb
Started weight (g)
Control
10
14.80
20.00
17.62 ± 1.96
100.00
  
VPA
10
14.90
18.80
17.47 ± 1.38
99.15
0.845
0.001
VPA-CUR
10
24.50
31.70
27.81 ± 2.82c
157.83
0.001
CUR
10
15.20
18.20
16.46 ± 1.04
93.42
0.121
Final weight (g)
Control
10
26.10
78.90
58.42 ± 21.45
100.00
 
0.001
VPA
10
17.20
75.40
42.21 ± 27.85
72.25
0.163
VPA-CUR
10
69.30
100.30
88.18 ± 12.10c
150.94
0.002
CUR
10
55.60
70.00
63.22 ± 5.61
108.22
0.509
Weight Gained
Control
10
11.30
58.90
40.80 ± 19.56
100.00
 
0.001
VPA
10
-0.30
60.30
24.74 ± 28.56
60.64
0.162
VPA-CUR
10
44.80
72.90
60.37 ± 9.80c
147.97
0.014
CUR
10
40.40
51.80
46.76 ± 4.80
114.61
0.371
Brain weight (g)
Control
10
1.17
1.61
1.38 ± 0.15
100.00
  
VPA
10
1.12
1.63
1.31 ± 0.17
95.08
0.361
0.001
VPA-CUR
10
1.51
1.72
1.61 ± 0.06c
116.54
0.001
CUR
10
1.29
1.59
1.43 ± 0.09
103.58
0.377
Opening eyes (Days)
Control
10
15.00
16.00
15.30 ± 0.48
100.00
 
0.001
VPA
10
10.00
17.00
14.50 ± 3.21
94.77
0.454
VPA-CUR
10
14.00
16.00
15.20 ± 1.03
99.35
0.786
CUR
10
11.00
13.00
12.0 0.47c
78.43
0.001
a P value between control group and other groups
b P value between all groups
cIndicates there is significant difference between the group and control at 0.05 level.
Table 2 exhibits the significant depletion of IFN-γ and non-significant depletion of 5HT and glutamine upon VPA exposure compared to the control group. CYP450 was significantly increased, while IL-6 and glutamate were non-significantly increased in VPA-exposed pups, and curcumin was effective in restoring nearly all the parameters, as shown in Table 2.
Table 2
Mean ± S.D. together with the independent t-test for Interleukin-6, Interferon Gamma, cytochrome P450, Serotonin, Glutamine, Glutamate together with Glutamate/Glutamine Ratio in neuro-intoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameter
Group
N
Min
Max
Mean ± S.D.
Percent Change
P valuea
P valueb
IL-6 (pg\ml)
Control
10
4.50
22.29
12.59 ± 6.07
100.00
 
0.019
VPA
10
3.61
24.96
15.47 ± 7.56
122.86
0.360
VPA-CUR
10
4.50
11.61
7.96 ± 2.22
63.25
0.044
CUR
10
3.61
16.95
9.87 ± 3.86
78.36
0.246
IFN-γ (pg\ml)
Control
10
206.47
407.59
331.64 ± 87.78
100.00
 
0.038
VPA
10
152.14
343.25
249.08 ± 58.88
75.10
0.024
VPA-CUR
10
167.86
366.84
244.41 ± 69.94c
73.70
0.024
CUR
10
117.82
359.21
237.26 ± 95.02c
71.54
0.033
CYP450 (ng\ml)
Control
10
33.02
40.16
36.46 ± 1.79
100.00
 
0.085
VPA
10
33.85
42.01
39.11 ± 2.64
107.27
0.017
VPA-CUR
10
34.62
43.83
37.50 ± 2.82
102.87
0.335
CUR
10
34.14
43.83
38.96 ± 2.92
106.85
0.033
5HT (ng\ml)
Control
10
113.92
169.46
148.75 ± 15.22
100.00
 
0.228
VPA
10
109.57
174.05
140.14 ± 22.04
94.22
0.323
VPA-CUR
10
85.18
181.05
135.77 ± 32.81
91.28
0.278
CUR
10
122.13
215.83
158.84 ± 30.84
106.78
0.366
Glutamine (pmol\ml)
Control
10
1640.31
2540.39
2186.87 ± 302.70
100.00
 
0.001
VPA
10
1861.71
2549.07
2082.25 ± 216.82
95.22
0.386
VPA-CUR
10
1456.53
2052.73
1763.15 ± 165.27c
80.62
0.002
CUR
10
1388.51
2324.78
1798.91 ± 274.83c
82.26
0.008
Glutamate (nmol\ml)
Control
10
215.20
286.01
249.53 ± 24.68
100.00
 
0.002
VPA
10
235.78
283.18
258.51 ± 14.04
103.60
0.334
VPA-CUR
10
178.70
283.58
232.05 ± 34.97
92.99
0.213
CUR
10
168.81
272.09
206.71 ± 39.12c
82.84
0.009
Glutamate/Glutamine Ratio
Control
10
0.10
0.16
0.116 ± 0.018
100.00
 
0.063
VPA
10
0.11
0.14
0.125 ± 0.011
108.04
0.169
VPA-CUR
10
0.10
0.16
0.132 ± 0.017
113.83
0.051
CUR
10
0.10
0.14
0.115 ± 0.016
99.65
0.963
a P value between control group and other groups
b P value between all groups
cIndicates there is significant difference between the group and control at 0.05 level
Table 3 shows lipid peroxide, oxidized glutathione, reduced glutathione, and glutathione S-transferases levels in all of the treated groups, along with the GSH/GSSG ratios. Non-significant decreases in LPO and GSTs in the VPA group were observed in all treated groups compared to the control group. The same table demonstrates the significant decrease in GSH in VPA and VPA-CUR compared to the control group. GSSG showed a significant increase in all of the treated pups compared with the control group. Table 4 and Fig. 2 present the Pearson’s correlations between the measured parameters.
Table 3
Mean ± S.D. together with the independent t-test for Lipid Peroxide, Oxidized Glutathione, Reduced Glutathione, Glutathione S-transferase together with GSH/GSSG ratio between neuro -intoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameter
Group
N
Min
Max
Mean ± S.D.
Percent Change
P valuea
P valueb
LPO (U\ml)
Control
10
2.70
5.00
3.68 ± 0.84
100.00
 
0.001
VPA
10
3.10
3.90
3.52 ± 0.24
95.65
0.573
VPA-CUR
10
1.40
3.40
2.46 ± 0.55c
66.73
0.001
CUR
10
1.40
3.70
2.56 ± 0.82c
69.57
0.007
GSSG (pg/ml)
Control
10
65.00
125.00
92.50 ± 23.24
100.00
 
0.001
VPA
10
150.00
310.00
225.00 ± 51.91c
243.24
0.001
VPA-CUR
10
225.00
460.00
345.00 ± 81.99c
372.97
0.001
CUR
10
100.00
180.00
138.50 ± 21.48
149.73
0.001
GSH (pg\ml)
Control
10
7104.06
9134.06
8016.97 ± 638.64
100.00
 
0.001
VPA
10
3506.25
5417.81
4397.66 ± 557.53c
54.85
0.001
VPA-CUR
10
4419.06
6040.94
5099.31 ± 485.93c
63.61
0.001
CUR
10
6825.00
9638.75
8190.53 ± 948.88
102.16
0.637
GSTs (mU\ml)
Control
10
8.50
20.50
15.35 ± 3.98
100.00
 
0.001
VPA
10
11.60
18.40
13.84 ± 2.00
90.19
0.304
VPA-CUR
10
8.50
17.00
12.74 ± 2.74
83.00
0.105
CUR
10
5.50
17.00
11.57 ± 3.78c
75.37
0.043
GSH/GSSG
Control
10
59.15
140.52
92.59 ± 27.69
100.00
 
0.001
VPA
10
13.87
31.68
20.54 ± 5.53c
22.19
0.001
VPA-CUR
10
9.61
22.22
15.64 ± 4.24c
16.89
0.001
CUR
10
47.43
87.58
60.33 ± 11.63c
65.16
0.005
a P value between control group and other groups
b P value between all groups
cIndicates there is significant difference between the group and control at 0.05 level
Table 4
Pearson Correlations between the measured parameters
Parameters
R (Person Correlation)
Sig.
 
Started weight (g) ~ Final weight (g)
0.641**
0.001
Pa
Started weight (g) ~ Weight Gained
0.494**
0.001
Pa
Started weight (g) ~ Brain weight (g)
0.652**
0.001
Pa
Started weight (g) ~ Opening eyes after (Days)
0.313*
0.049
Pa
Started weight (g) ~ Glutamate/Glutamin Ratio
0.322*
0.043
Pa
Started weight (g) ~ GSH (mmol/L)
−0.438**
0.005
Nb
Started weight (g) ~ GSSG (pg/ml)
0.765**
0.001
Pa
Started weight (g) ~ GSH (pg\ml)
−0.438**
0.005
Nb
Started weight (g) ~ GSH + GSSG
-0.408**
0.009
Nb
Started weight (g) ~ GSH/GSSG
-0.479**
0.002
Nb
Final weight (g) ~ Weight Gained
0.984**
0.001
Pa
Final weight (g) ~ Brain weight (g)
0.887**
0.001
Pa
Final weight (g) ~ LPO (U\ml)
−0.371*
0.019
Nb
Final weight (g) ~ GSSG (pg/ml)
0.390*
0.013
Pa
Weight Gained ~ Brain weight (g)
0.853**
0.001
Pa
Weight Gained ~ LPO (U\ml)
−0.353*
0.025
Nb
Brain weight (g) ~ LPO (U\ml)
−0.313*
0.049
Nb
Brain weight (g) ~ GSSG (pg/ml)
0.369*
0.019
Pa
Opening eyes after (Days) ~ GSH (mmol/L)
−0.326*
0.04
Nb
Opening eyes after (Days) ~ GSH (pg\ml)
−0.326*
0.04
Nb
Opening eyes after (Days) ~ GSH + GSSG
-0.322*
0.043
Nb
IL-6 (pg\ml) ~ Glutamin (pmol\ml)
0.377*
0.017
Pa
IL-6 (pg\ml) ~ Glutamate (nmol\ml)
0.322*
0.043
Pa
IL-6 (pg\ml) ~ LPO (U\ml)
0.318*
0.045
Pa
IFN-g (pg\ml) ~ GSTs (mU\ml)
0.588**
0.001
Pa
IFN-g (pg\ml) ~ Glutamin (pmol\ml)
0.541**
0.001
Pa
IFN-g (pg\ml) ~ Glutamate (nmol\ml)
0.502**
0.001
Pa
IFN-g (pg\ml) ~ LPO (U\ml)
0.523**
0.001
Pa
CYP450 (ng\ml) ~ GSTs (mU\ml)
−0.347*
0.028
Nb
5HT (ng\ml) ~ GSSG (pg/ml)
−0.352*
0.026
Nb
GSTs (mU\ml) ~ Glutamin (pmol\ml)
0.597**
0.001
Pa
GSTs (mU\ml) ~ Glutamate (nmol\ml)
0.451**
0.004
Pa
GSTs (mU\ml) ~ LPO (U\ml)
0.650**
0.001
Pa
Glutamin (pmol\ml) ~ Glutamate (nmol\ml)
0.635**
0.001
Pa
Glutamin (pmol\ml) ~ Glutamate/Glutamin Ratio
−0.414**
0.008
Nb
Glutamin (pmol\ml) ~ LPO (U\ml)
0.722**
0.001
Pa
Glutamate (nmol\ml) ~ Glutamate/Glutamin Ratio
0.435**
0.005
Pa
Glutamate (nmol\ml) ~ LPO (U\ml)
0.678**
0.001
Pa
Glutamate/Glutamin Ratio ~ GSSG (pg/ml)
0.337*
0.033
Pa
GSH (mmol/L) ~ GSSG (pg/ml)
−0.684**
0.001
Nb
GSH (mmol/L) ~ GSH (pg\ml)
1.000**
0.001
Pa
GSH (mmol/L) ~ GSH + GSSG
0.999**
0.001
Pa
GSH (mmol/L) ~ GSH/GSSG
0.819**
0.001
Pa
GSSG (pg/ml) ~ GSH (pg\ml)
−0.684**
0.001
Nb
GSSG (pg/ml) ~ GSH + GSSG
-0.651**
0.001
Nb
GSSG (pg/ml) ~ GSH/GSSG
-0.816**
0.001
Nb
GSH (pg\ml) ~ GSH + GSSG
0.999**
0.001
Pa
GSH (pg\ml) ~ GSH/GSSG
0.819**
0.001
Pa
GSH + GSSG ~ GSH/GSSG
0.803**
0.001
Pa
**Correlation is significant at the 0.01 level.
*Correlation is significant at the 0.05 level.
aPositive Correlation.
bNegative Correlation
Receiver operating characteristics curves are presented in Fig. 3. Area under the curve (AUC), cutoff values, sensitivity and specificity are listed in Tables 5, 6 and 7
Table 5
ROC Curve of weight gain, brain weight and age of eye opening between neuro-intoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameters
Groups
Area under the curve
Cutoff value
Sensitivity %
Specificity %
Started weight (g)
VPA
0.600
18.900
100.0%
40.0%
VPA-CUR
1.000
22.250
100.0%
100.0%
CUR
0.670
18.250
100.0%
60.0%
Final weight (g)
VPA
0.700
25.450
60.0%
100.0%
VPA-CUR
0.915
77.550
80.0%
90.0%
CUR
0.585
71.300
100.0%
40.0%
Weight Gained
VPA
0.640
8.700
60.0%
100.0%
VPA-CUR
0.800
60.200
60.0%
100.0%
CUR
0.530
52.700
100.0%
40.0%
Brain weight (g)
VPA
0.640
1.415
80.0%
50.0%
VPA-CUR
0.905
1.477
100.0%
80.0%
CUR
0.590
1.364
90.0%
40.0%
Opening eyes after (Days)
VPA
0.570
16.500
50.0%
100.0%
VPA-CUR
0.510
15.500
60.0%
70.0%
CUR
1.000
14.000
100.0%
100.0%
Table 6
ROC Curve of Interleukin-6, Interferon Gamma, cytochrome P450, Serotonin, Glutamine, Glutamate together with Glutamate/Glutamine ratio in neuro-intoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameters
Groups
Area under the curve
Cutoff value
Sensitivity %
Specificity %
IL-6 (pg\ml)
VPA
0.625
9.832
80.0%
40.0%
VPA-CUR
0.740
11.165
90.0%
60.0%
CUR
0.635
12.945
80.0%
50.0%
IFN-γ (pg\ml)
VPA
0.750
318.225
90.0%
70.0%
VPA-CUR
0.780
327.520
90.0%
70.0%
CUR
0.805
365.885
100.0%
60.0%
CYP450 (ng\ml)
VPA
0.790
37.085
80.0%
80.0%
VPA-CUR
0.590
37.471
50.0%
90.0%
CUR
0.790
37.630
80.0%
90.0%
5HT (ng\ml)
VPA
0.620
137.220
60.0%
90.0%
VPA-CUR
0.625
140.965
60.0%
80.0%
CUR
0.550
157.750
50.0%
80.0%
GSTs (mU\ml)
VPA
0.620
17.100
90.0%
40.0%
VPA-CUR
0.675
12.450
50.0%
90.0%
CUR
0.730
12.450
60.0%
90.0%
Glutamin (pmol\ml)
VPA
0.610
2365.295
90.0%
40.0%
VPA-CUR
0.890
1984.715
90.0%
80.0%
CUR
0.840
2007.865
90.0%
80.0%
Glutamate (nmol\ml)
VPA
0.590
244.955
90.0%
50.0%
VPA-CUR
0.645
234.870
70.0%
70.0%
CUR
0.810
213.790
70.0%
100.0%
Glutamate/Glutamin ratio
VPA
0.810
0.111
100.0%
60.0%
VPA-CUR
0.820
0.122
80.0%
90.0%
CUR
0.500
0.115
60.0%
70.0%
Table 7
ROC Curve of Lipid Peroxide, Oxidized Glutathione, Reduced Glutathione, Glutathione S-transferase together with GSH/GSSG ratio between neuro-intoxicated, protected and therapeutically treated rat pups compared to healthy control
Parameters
Groups
Area under the curve
Cutoff value
Sensitivity %
Specificity %
LPO (U\ml)
VPA
0.545
3.250
90.0%
50.0%
VPA-CUR
0.915
2.650
80.0%
100.0%
CUR
0.805
3.150
80.0%
70.0%
GSH (mmol/L)
VPA
1.000
200.350
100.0%
100.0%
VPA-CUR
1.000
210.320
100.0%
100.0%
CUR
0.570
278.195
40.0%
90.0%
GSSG (pg/ml)
VPA
1.000
137.500
100.0%
100.0%
VPA-CUR
1.000
175.000
100.0%
100.0%
CUR
0.935
117.500
90.0%
90.0%
GSH (pg\ml)
VPA
1.000
6260.938
100.0%
100.0%
VPA-CUR
1.000
6572.500
100.0%
100.0%
CUR
0.570
8693.594
40.0%
90.0%
GSH + GSSG
VPA
1.000
6435.938
100.0%
100.0%
VPA-CUR
1.000
6762.500
100.0%
100.0%
CUR
0.580
8798.594
40.0%
90.0%
GSH/GSSG
VPA
1.000
45.417
100.0%
100.0%
VPA-CUR
1.000
40.685
100.0%
100.0%
CUR
0.900
71.255
90.0%
80.0%

Discussion

Prenatal VPA exposure resulted in delayed maturation in newborns, as evidenced by lower body weight, a slight decrease in brain weight and late eye opening, indicating some altered neurodevelopmental effects. Our results validate many previous studies that suggest there is a maturational delay in the early stage of life of VPA-exposed rats [17]. Significant effects of postnatal curcumin treatments were found, that ameliorated all of the observed development delays in the current study. These results indicate the therapeutic potential of curcumin as a neuroprotective agent in the treatment of delayed maturation. Our results are consistent with many recent studies on discovering redevelopment and accelerating motor functional recovery of curcumin treatment in mice [18].
Elevated levels of IL-6 in the CNS have been reported in a number of neurological diseases that are associated with brain injury or inflammation [19]. In the current study, VPA exposure amplified the level of IL-6 in the brain tissue, which may be due to neuroinflammation and altering the immune response during brain development. However, curcumin treatment was able to decrease IL-6 levels, as curcumin can suppress the pro-inflammatory gene expression by blocking phosphorylation of the inhibitory factor I-kappa B (IκB) [20]. VPA is generally used in the treatment of epilepsy, but recently, it has been found to be effective in the treatment of oncolytic herpes simplex virus (oHSV) infection, as this drug can inhibit the expression of IFN-β and the IFN-mediated proteins STAT1 and PKR in infected cells [21]. This could support our results showing a significant decrease of IFN-ɣ in VPA-exposed pups. Additionally, the remarkable decrease in this parameter in curcumin-treated pups may be due to the anti-inflammatory action of curcumin, which causes inhibition of production of cytokines, such as interferon-ɣ, due to suppression of the Janus kinase (JAK)-STAT signaling cascade [22]. Cytochrome P450 was found to be increased in VPA-exposed pups while a slight decrease was observed in VPA-CUR and CUR groups, as shown in Table 2. The cytochrome P450 enzymes are the major catalysts involved in the metabolism of many psychoactive drugs in the brain. The significant increase in CYT-P450 in VPA-exposed pups could be due to the availability of VPA as a substrate. Curcumin, a good antioxidant, was slightly effective in decreasing the activity of CYT-P450 [23]. The role of CYT-P450 in the metabolism of VPA can also be explained through non-significant changes in the concentration of lipid peroxides as markers of oxidative stress, as demonstrated in Table 3. The same table demonstrates the antioxidant effects of curcumin, as shown by significant decreases of lipid peroxides in VPA-CUR and CUR groups compared to the VPA group. An unexpected finding of the present study is that VPA does not induce elevation of lipid peroxides as markers of oxidative stress. This finding could be attributed to the fact that VPA is an anti-epileptic drug that is designed to have the least toxic effects on treated patients. The significant decrease in lipid peroxide levels in VPA-CUR and CUR groups compared to the control group is consistent with various models posed by several authors, which proves that curcumin is a good antioxidant that inhibits lipid peroxidation [24].
Glutathione-S-transferases (GSTs) play a key role in enzymatic detoxification and were found to be 10.19% lower in the VPA group than in the control group (Table 3), which is due to the neurotoxic effect of VPA through insufficient conjugation of electrophiles and detoxification of the reactive species, as previously described by Chaudhary & Parvez, in the cerebellum and cerebral cortex of the rat brain [25]. Rats fed dietary curcumin were found to have decreased hepatic GST activity compared to controls because of competitive enzyme inhibition by the curcumin molecule [26]. This could explain the remarkable decrease of GST in the VPA-CUR and CUR groups.
Table 2 shows the non-significant decreased level of serotonin in the VPA group compared to the control group. On the other hand, an increase in serotonin in the CUR group was observed compared to the control, the VPA and the VPA-CUR groups, with values of 6.78, 12.56 and 15.5%, respectively. This is not consistent with a previous study, in which increased levels of serotonin were found in the brains of rats that had been prenatally exposed to VPA in association with disrupted sleep/awake rhythms [27]. Aside from the well-known deficiency in serotonergic neurotransmission as a pathophysiological correlate of autism, recent evidence points to the pivotal role of increased glutamate receptor activation as well. While the present study demonstrates a non-significant decrease in brain serotonin of VPA-exposed rats, a remarkable elevation in brain glutamate was recorded. A hypothesis integrating current concepts of neurotransmission and hypothalamus-pituitary-adrenal (HPA) axis dysregulation with findings on immunological alterations was proposed by Müller and Schwarz [28]. Immune activation, including increased production of pro-inflammatory cytokines, has repeatedly been described in mild depression. Pro-inflammatory cytokines such as IL-2 and IL-6 activate the tryptophan- and serotonin-degrading enzyme indole amine 2,3-dioxygenase (IDO). Based on this hypothesis, the increase in IL-6 reported in the present study can be related to the decrease in serotonin levels. A VPA-exposed developmental rodent model in the present study may show persistent autistic features that present biochemically as low serotonin and high glutamate levels [29].
Glutamate is an excitatory neurotransmitter that is usually transported from neurons to astrocytes in order to be buffered through the formation of glutamine; the glutamate/glutamine ratio can be a useful marker for the decrease of excitotoxicity. Table 3 shows the non-significant elevation of glutamate along with the unchanged glutamine and glutamate/glutamine ratio in the VPA-exposed rats compared to the control group. This is supported by the previous work of Bristot Silvestrin et al. [30], which reported unaltered glutamate uptake in 15 day old rats that were prenatally exposed to VPA and showed a 160% increase at an age of 120 days. The anti-excitotoxicity effect of curcumin, presented as a much lower glutamate level in cur-treated rats compared to VPA-exposed rats, and even the control group, can be easily related to its protective effect against glutamate excitotoxicity [31].
GSH was significantly lower in VPA-exposed rat pups (7 days old), compared to the control group (Table 3) This is consistent with the recent study by Bristot Silvestrin et al. [30], which recorded unaltered GSH levels in 15-day-old rats that were prenatally exposed to VPA. This can be related to the non-significant elevation in glutamate reported in the present study. The unchanged GSH and glutamate levels recorded in the present study do not contradict the use of VPA-exposed rats as rodent models of autism. This opinion can be supported with the previously mentioned significant impairment of both parameters in 120-day-old rats that were exposed to VPA during pregnancy. The neurotoxic effect of VPA, along with the neuro-therapeutic and antioxidant effects of curcumin can be observed together in Table 3.9. A highly significant decrease in GSSG in VPA-exposed rats reflects the impairment of total antioxidant and glutathione status in this group of animals. GSSG, as an oxidized form of glutathione, can be easily converted to GSH by glutathione reductase, and hence, a lower concentration can easily lead to low GSH. In our study, VPA-exposed rats were under stressful conditions, so the unexpected increase of GSSG in valproate-treated animals that were also treated with curcumin, is supported by the previous study by Hagl et al. [32], who reported that when under non-stressful conditions, curcumin induces the synthesis of GSH and many detoxifying enzymes (as shown in group IV). This might also be attributed to the low absorption and quick elimination of curcumin from the body. Hagl et al. proved that low bioavailability of curcumin can be ameliorated through administration with secondary plant compounds, micronization and micellation, which might help to increase its therapeutic potency [32].
Tables 5, 6 and 7 present the ROC curve parameters of all measured variables from all test groups. It is readily apparent that while some parameters show effectiveness as biomarkers for VPA neurotoxicity, others are good to excellent markers for CUR therapeutic and/or antioxidant effects. The postnatal growth and maturation markers presented by weight gain, brain weight and eye opening demonstrated AUC ranges between 0.7 and 1, which support their use as markers for VPA neurotoxicity and curcumin therapeutic potency. IL-6 shows smaller AUC values compared to IFN-γ, which suggests that the latter is a good marker for VPA toxicity and curcumin therapeutic and antioxidant effects. Lipid peroxides and CYT P450 both demonstrate a satisfactory AUC, which shows that both can be used to test the efficacy of the curcumin antioxidant effect. Serotonin, GST, glutamate and glutamine all showed relatively poor potency as markers for VPA neurotoxicity and were good markers for curcumin efficacy. Conversely, GSSG and GSH both show a high predictive value that can be used as markers of VPA neurotoxicity and of curcumin’s therapeutic effect. The increase in GSSG in valproate-treated animals that were also treated with curcumin was unexpected but is supported in the previous study by Reyes et al. (2013), who reported that under non-stress conditions, curcumin induces the synthesis of GSH and of many detoxifying and cytoprotective enzymes. Based on this, our data suggest that pretreatment with curcumin may have more protective effects than therapeutic antioxidant effects but still demonstrates therapeutic effects in ameliorating IL-6, INF-ɣ, LOP, GST,5-HT and GSH.

Conclusion

To summarize, this study shows evidence of the postnatal therapeutic role of curcumin in improving most of the impaired parameters in VPA-induced rodent models with persistent autistic features. The mechanism of action underlying the therapeutic effects of curcumin should be investigated in the near future. Studies of the protective effects of curcumin are also recommended.

Acknowledgments

This research project was supported by a grant from the research center of the Center for Female Scientific and Medical Colleges at King Saud University.

Funding

Research center of the Center for Female Scientific and Medical Colleges at King Saud University.

Availability of data and material

Data is available as supplementary excel sheet (Additional file 1).

Authors’ contributions

MA: Performed the practical work; RS: Co-drafted the manuscript and designed the illustrated experimental chart; MS: Supervised the experimental work related to VPA dosage; LA: Revised the manuscript and did the statistical analysis; AE: Suggested the topic and co-drafted the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
This work was approved by the Ethical Committee of Science College at King Saud University (approval no KSU-IRB008E.)

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Ergaz Z, Weinstein-Fudim L, Ornoy A. Genetic and non-genetic animal models for autism spectrum disorders (ASD) Reprod. Toxicology. 2016;64:116–40. Ergaz Z, Weinstein-Fudim L, Ornoy A. Genetic and non-genetic animal models for autism spectrum disorders (ASD) Reprod. Toxicology. 2016;64:116–40.
3.
Zurück zum Zitat Kazlauskas N, Campolongo M, Lucchina L, Zappala C, Depino AM. Postnatal behavioral and inflammatory alterations in female pupsprenatally exposed to valproic acid. Psychoneuroendocrinology. 2016;72:11–21. Kazlauskas N, Campolongo M, Lucchina L, Zappala C, Depino AM. Postnatal behavioral and inflammatory alterations in female pupsprenatally exposed to valproic acid. Psychoneuroendocrinology. 2016;72:11–21.
4.
Zurück zum Zitat Kim JW, Seung H, Kim KC, Gonzales EL, Oh HA, Yang SM, Ko MJ, Han SH, Banerjee S, Shin CY. Agmatine rescues autistic behaviors in the valproic acid-induced animal model of autism. Neuropharmacology. 2017;113(Pt A):71–81. Kim JW, Seung H, Kim KC, Gonzales EL, Oh HA, Yang SM, Ko MJ, Han SH, Banerjee S, Shin CY. Agmatine rescues autistic behaviors in the valproic acid-induced animal model of autism. Neuropharmacology. 2017;113(Pt A):71–81.
5.
Zurück zum Zitat Christensen J, Grønborg TK, Sørensen MJ, Schendel D, Parner ET, Pedersen LH, Vestergaard M. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309(16):1696–703.CrossRefPubMedPubMedCentral Christensen J, Grønborg TK, Sørensen MJ, Schendel D, Parner ET, Pedersen LH, Vestergaard M. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309(16):1696–703.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Kim KC, Kim P, Go HS, Choi CS, Yang SI, Cheong JH, Shin CY, Ko KH. The critical period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats. Toxicol Lett. 2011;201:137–42.CrossRefPubMed Kim KC, Kim P, Go HS, Choi CS, Yang SI, Cheong JH, Shin CY, Ko KH. The critical period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats. Toxicol Lett. 2011;201:137–42.CrossRefPubMed
7.
Zurück zum Zitat Ghodke-Puranik Y, Thorn CF, Lamba JK, Leeder JS, Song W, Birnbaum AK, Klein TE. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics. 2013;23(4):236–41.CrossRefPubMedPubMedCentral Ghodke-Puranik Y, Thorn CF, Lamba JK, Leeder JS, Song W, Birnbaum AK, Klein TE. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics. 2013;23(4):236–41.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Johannessen CU, Johannessen SI. Valproate: past, present, and future. CNS Drug Rev. 2003;9:199–216.CrossRefPubMed Johannessen CU, Johannessen SI. Valproate: past, present, and future. CNS Drug Rev. 2003;9:199–216.CrossRefPubMed
9.
Zurück zum Zitat Bromley RL, Baker GA, Meador KJ. Cognitive abilities and behaviour of children exposed to antiepileptic drugs in utero. Curr Opin Neurol. 2009;22:162–6.CrossRefPubMedPubMedCentral Bromley RL, Baker GA, Meador KJ. Cognitive abilities and behaviour of children exposed to antiepileptic drugs in utero. Curr Opin Neurol. 2009;22:162–6.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat McVearry KM, Gaillard WD, VanMeter J, Meador KJ. A prospective study of cognitive fluency and originality in children exposed in utero to carbamazepine, lamotrigine, or valproate monotherapy. Epilepsy Behav. 2009;16:609–16.CrossRefPubMedPubMedCentral McVearry KM, Gaillard WD, VanMeter J, Meador KJ. A prospective study of cognitive fluency and originality in children exposed in utero to carbamazepine, lamotrigine, or valproate monotherapy. Epilepsy Behav. 2009;16:609–16.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Meador KJ, Baker GA, Browning N, Cohen MJ, Clayton-Smith J, Kalayjian LA, Kanner A, Liporace JD, Pennell PB, Privitera M, Loring DW. NEAD Study Group Foetal antiepileptic drug exposure and verbal versus non-verbal abilities at three years of age. Brain. 2011;134(Pt 2):396–404.CrossRefPubMedPubMedCentral Meador KJ, Baker GA, Browning N, Cohen MJ, Clayton-Smith J, Kalayjian LA, Kanner A, Liporace JD, Pennell PB, Privitera M, Loring DW. NEAD Study Group Foetal antiepileptic drug exposure and verbal versus non-verbal abilities at three years of age. Brain. 2011;134(Pt 2):396–404.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Mythri RB, Bharath MM. Curcumin: a potential neuroprotective agent in Parkinson's disease. Curr Pharm Des. 2012;18(1):91–9.CrossRefPubMed Mythri RB, Bharath MM. Curcumin: a potential neuroprotective agent in Parkinson's disease. Curr Pharm Des. 2012;18(1):91–9.CrossRefPubMed
13.
Zurück zum Zitat Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases Int. J Biochem Cell Biol. 2009;41(1):40–59.CrossRef Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases Int. J Biochem Cell Biol. 2009;41(1):40–59.CrossRef
14.
Zurück zum Zitat Bhandari R. Kuhad a Neuropsychopharmacotherapeutic efficacy of curcumin in experimental paradigm of autism spectrum disorders. Life Sci. 2015;141:156–69.CrossRefPubMed Bhandari R. Kuhad a Neuropsychopharmacotherapeutic efficacy of curcumin in experimental paradigm of autism spectrum disorders. Life Sci. 2015;141:156–69.CrossRefPubMed
15.
Zurück zum Zitat Schneider T, Przewłocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30(1):80–9.CrossRefPubMed Schneider T, Przewłocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30(1):80–9.CrossRefPubMed
16.
Zurück zum Zitat Purwar B, Shrivastava A, Arora N, Saxena AKAY. Effects of Curcumin on the gastric emptying of albino rats. Indian J Physiol Pharmacol. 2012;56(2):168–73.PubMed Purwar B, Shrivastava A, Arora N, Saxena AKAY. Effects of Curcumin on the gastric emptying of albino rats. Indian J Physiol Pharmacol. 2012;56(2):168–73.PubMed
17.
Zurück zum Zitat Roullet FI, Lai JK, Foster JA. In utero exposure to valproic acid and autism–a current review of clinical and animal studies. Neurotoxicol Teratol. 2013;36:47–56.CrossRefPubMed Roullet FI, Lai JK, Foster JA. In utero exposure to valproic acid and autism–a current review of clinical and animal studies. Neurotoxicol Teratol. 2013;36:47–56.CrossRefPubMed
18.
Zurück zum Zitat Ma J, Liu J, Yu H, Wang Q, Chen Y, Xiang L. Curcumin promotes nerve regeneration and functional recovery in rat model of nerve crush injury. Neurosci Lett. 2013;547:26–31.CrossRefPubMed Ma J, Liu J, Yu H, Wang Q, Chen Y, Xiang L. Curcumin promotes nerve regeneration and functional recovery in rat model of nerve crush injury. Neurosci Lett. 2013;547:26–31.CrossRefPubMed
20.
Zurück zum Zitat Jurenka JS. Anti-inflammatory properties of Curcumin, a major constituent of Curcuma longa: a review of preclinical and clinical research. Altern Med Rev. 2009;14(2):141–53.PubMed Jurenka JS. Anti-inflammatory properties of Curcumin, a major constituent of Curcuma longa: a review of preclinical and clinical research. Altern Med Rev. 2009;14(2):141–53.PubMed
21.
Zurück zum Zitat Otsuki A, et al. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol Ther. 2008;16:1546–55.CrossRef Otsuki A, et al. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol Ther. 2008;16:1546–55.CrossRef
22.
Zurück zum Zitat Kim HY, Park EJ, Joe EH, et al. Curcumin suppresses januskinase-STAT inflammatory signalling through activation of Srchomology 2 domain-containing tyrosine phosphatase 2 in brain microglia. J Immunol. 2003;171:6072–9.CrossRefPubMed Kim HY, Park EJ, Joe EH, et al. Curcumin suppresses januskinase-STAT inflammatory signalling through activation of Srchomology 2 domain-containing tyrosine phosphatase 2 in brain microglia. J Immunol. 2003;171:6072–9.CrossRefPubMed
23.
Zurück zum Zitat Sharma RA, Gescher AJ, Steward WP. Curcumin: the story so far. Eur J Cancer. 2005;41:1955–68.CrossRefPubMed Sharma RA, Gescher AJ, Steward WP. Curcumin: the story so far. Eur J Cancer. 2005;41:1955–68.CrossRefPubMed
24.
Zurück zum Zitat El-Demerdasha FM, Yousef MI, Radwan FM. Ameliorating effect of curcumin on sodium arsenite-induced oxidative damage and lipid peroxidation in different rat organs. Food Chem Toxicol. 2009;47(1):249–54.CrossRef El-Demerdasha FM, Yousef MI, Radwan FM. Ameliorating effect of curcumin on sodium arsenite-induced oxidative damage and lipid peroxidation in different rat organs. Food Chem Toxicol. 2009;47(1):249–54.CrossRef
25.
Zurück zum Zitat Chaudhary S, Parvez S. An in vitro approach to assess the neurotoxicityof valproic acid-induced oxidative stress in cerebellumand cerebral cortex of young rats. Neuroscience. 2012;225:258–68.CrossRefPubMed Chaudhary S, Parvez S. An in vitro approach to assess the neurotoxicityof valproic acid-induced oxidative stress in cerebellumand cerebral cortex of young rats. Neuroscience. 2012;225:258–68.CrossRefPubMed
26.
Zurück zum Zitat Piper JT, Singhal SS, Salameh M, Torman RT, Awasthi YC, Awasthi S. Mechanisms of anticarcinogenic properties ofcurcumin: the effect of curcumin on glutathione linked detoxificationenzymes in rat liver. Int J Biochem Cell Biol. 1998;30:445–56.CrossRefPubMed Piper JT, Singhal SS, Salameh M, Torman RT, Awasthi YC, Awasthi S. Mechanisms of anticarcinogenic properties ofcurcumin: the effect of curcumin on glutathione linked detoxificationenzymes in rat liver. Int J Biochem Cell Biol. 1998;30:445–56.CrossRefPubMed
27.
Zurück zum Zitat Gottfried C, Bambini-Junior V, Baronio D, Zanatta G, Silvestrin RB, Vaccaro T, Riesgo R. In: Fitzgerald M, editor. ISBN: 978-953-51-1021-7, In Tech Valproic acid in autism spectrum disorder: from an environmental risk factor to a reliable animal model, recent advances in autism spectrum disorders, vol. 1; 2013. doi:10.5772/54824. Gottfried C, Bambini-Junior V, Baronio D, Zanatta G, Silvestrin RB, Vaccaro T, Riesgo R. In: Fitzgerald M, editor. ISBN: 978-953-51-1021-7, In Tech Valproic acid in autism spectrum disorder: from an environmental risk factor to a reliable animal model, recent advances in autism spectrum disorders, vol. 1; 2013. doi:10.​5772/​54824.
28.
Zurück zum Zitat Müller N, Schwarz MJ. The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol Psychiatry. 2007;12(11):988–1000.CrossRefPubMed Müller N, Schwarz MJ. The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol Psychiatry. 2007;12(11):988–1000.CrossRefPubMed
29.
Zurück zum Zitat Anderson G, Maes M. Redox regulation and the autistic Spectrum: role of tryptophan Catabolites, Immuno-inflammation, Autoimmunity and the Amygdala. Curr Neuropharmacol. 2014;12(2):148–67.CrossRefPubMedPubMedCentral Anderson G, Maes M. Redox regulation and the autistic Spectrum: role of tryptophan Catabolites, Immuno-inflammation, Autoimmunity and the Amygdala. Curr Neuropharmacol. 2014;12(2):148–67.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Bristot Silvestrin R, Bambini-Junior V, Galland F, Daniele Bobermim L, Quincozes-Santos A, Torres Abib R, Zanotto C, et al. Animal model of autism induced by prenatal exposure to valproate: altered glutamate metabolism in the hippocampus. Brain Res. 2013;1495:52–60.CrossRefPubMed Bristot Silvestrin R, Bambini-Junior V, Galland F, Daniele Bobermim L, Quincozes-Santos A, Torres Abib R, Zanotto C, et al. Animal model of autism induced by prenatal exposure to valproate: altered glutamate metabolism in the hippocampus. Brain Res. 2013;1495:52–60.CrossRefPubMed
31.
Zurück zum Zitat Wang R, Li YB, Li YH, Xu Y, Wu HL, Li XJ. Curcumin protects against glutamate excitotoxicity in rat cerebral cortical neurons by increasing brain-derived neurotrophic factor level and activating TrkB. Brain Res. 2008;1210:84–91.CrossRefPubMed Wang R, Li YB, Li YH, Xu Y, Wu HL, Li XJ. Curcumin protects against glutamate excitotoxicity in rat cerebral cortical neurons by increasing brain-derived neurotrophic factor level and activating TrkB. Brain Res. 2008;1210:84–91.CrossRefPubMed
32.
Zurück zum Zitat Hagl S, Kocher A, Schiborr C, Kolesova N, Frank J, Eckert GP. Curcumin micelles improve mitochondrial function in neuronal PC12 cells and brains of NMRI mice - impact on bioavailability. Neurochem Int. 2015;89:234–42.CrossRefPubMed Hagl S, Kocher A, Schiborr C, Kolesova N, Frank J, Eckert GP. Curcumin micelles improve mitochondrial function in neuronal PC12 cells and brains of NMRI mice - impact on bioavailability. Neurochem Int. 2015;89:234–42.CrossRefPubMed
Metadaten
Titel
Postnatal treatment using curcumin supplements to amend the damage in VPA-induced rodent models of autism
verfasst von
Maha Al-Askar
Ramesa Shafi Bhat
Manar Selim
Laila Al-Ayadhi
Afaf El-Ansary
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2017
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-017-1763-7

Weitere Artikel der Ausgabe 1/2017

BMC Complementary Medicine and Therapies 1/2017 Zur Ausgabe