Hostname: page-component-76fb5796d-wq484 Total loading time: 0 Render date: 2024-04-28T03:20:13.364Z Has data issue: false hasContentIssue false

In vitro screening of neuroprotective activity of Indian medicinal plant Withania somnifera

Published online by Cambridge University Press:  18 October 2017

Manjeet Singh*
Affiliation:
INRS – Institut Armand Frappier, 531, boul. des Prairies, Laval, Québec, Canada H7V 1B7
Charles Ramassamy
Affiliation:
INRS – Institut Armand Frappier, 531, boul. des Prairies, Laval, Québec, Canada H7V 1B7 Department of Medical Biology, Faculty of Medicine, Laval University, Québec, Canada G1K 7P4
*
* Corresponding author: Dr M. Singh, email msinghmore@gmail.com

Abstract

Canine cognitive dysfunction (CCD) is an age-dependent neurodegenerative condition characterised by changes in decline in learning and memory patterns. The neurodegenerative features of CCD in ageing dogs and cats are similar to human ageing and Alzheimer's disease (AD). Discovering neuroprotective disease-modifying therapies against CCD and AD is a major challenge. Strong evidence supports the role of amyloid β peptide deposition and oxidative stress in the pathophysiology of CCD and AD. In both the human and canine brain, oxidative damage progressively increases with age. Dietary antioxidants from natural sources hold a great promise in halting the progression of CCD and AD. Withania somnifera (WS), an Ayurvedic tonic medicine, also known as ‘Indian ginseng’ or ashwagandha has a long history of use in memory-enhancing therapy but there is a dearth of studies on its neuroprotective effects. The objective of this study was to investigate whether WS extract can protect against Aβ peptide- and acrolein-induced toxicity. We demonstrated that treatment with WS extract significantly protected the human neuroblastoma cell line SK-N-SH against Aβ peptide and acrolein in various cell survival assays. Furthermore, treatment with WS extract significantly reduced the generation of reactive oxygen species in SK-N-SH cells. Finally, our results showed that WS extract is also a potent inhibitor of acetylcholinesterase activity. Thus, our initial findings indicate that WS extract may act as an antioxidant and cholinergic modulator and may have beneficial effects in CCD and AD therapy.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s) 2017

Canine cognitive dysfunction (CCD) and Alzheimer's disease (AD) are the most common age-related neurodegenerative disorders affecting millions of pets and people worldwide. It is now well established that both oxidative stress and Aβ peptide have been implicated in the pathogenesis of CCD and AD( Reference Schütt, Helboe and Pedersen 1 ). Aβ peptide is formed and released after the sequential cleavage of the amyloid precursor protein by β- and γ-secretases, respectively. Aβ peptide, a 40–42 amino acid-long peptide, is an important component of senile plaque formation in AD brain( Reference Mattson 2 ) and its toxicity is mediated through the generation of hydrogen peroxide (H2O2)( Reference Behl, Davis and Lesley 3 , Reference Opazo, Huang and Cherny 4 ). Reactive oxygen species (ROS), on the other hand, generate highly electrophilic α,β-unsaturated carbonyl derivatives including acrolein, 4-hydroxy-2-nonenal, and 4-oxononenal from the peroxidation of membrane lipids( Reference LoPachin, Gavin and Petersen 5 ). In AD brain, levels of acrolein were found to be significantly higher in several brain regions such as in the hippocampus, amygdala, middle temporal gyrus and cerebellum( Reference Williams, Lynn and Markesbery 6 , Reference Bradley, Markesbery and Lovell 7 ). In primary neuronal cultures from the hippocampus, the toxicity of acrolein was higher than 4-hydroxy-2-nonenal( Reference Lovell, Xie and Markesbery 8 ). We have recently established on a neuronal cell line SK-N-SH the details of the multiprocess step of the toxicity of acrolein( Reference Thanh Nam, Arseneault and Zarkovic 9 ). We showed that, in addition to being a byproduct of lipid peroxidation, acrolein could also potentiate oxidative damage and activate several redox-sensitive pathways.

Incidence rates of age-specific AD in rural India are at least three times lower than those of an age-matched American reference population( Reference Chandra, Pandav and Dodge 10 ), which could be attributed to genetic, environmental or dietary causes. Withania somnifera (WS) is one of the most important medicinal plants used in the Indian system of traditional medicine as a nootropic agent and brain tonic to restore age-related decline in mental abilities( Reference Ven Murthy, Ranjekar and Ramassamy 11 ). The major biochemical constituents of WS are steroidal alkaloids and steroidal lactones saponins, together called withanolides( Reference Elsakka, Grigorescu and Stănescu 12 ). Presently more than twelve alkaloids and forty withanolides have been identified and characterised from the leaves, roots and berries of Withania species which either exist in free form, i.e. withanone, withaferin A, withanolide A, withanolides D-M, withanolides I–III, or in glycosidic form, i.e. withanosides I–VI( Reference Mirjalili, Moyano and Bonfill 13 ). Recently WS extract has been shown to protect against Aβ peptide- and H2O2-induced toxicities( Reference Kuboyama, Tohda and Komatsu 14 Reference Kumar, Seal and Howes 16 ). In spite of its widespread use as a brain tonic, its neuroprotective activity against acrolein remains poorly investigated.

Since multi-factorial causes have been recognised in CCD and AD as well as in other neurodegenerative disorders, CCD and AD will require multiple drug therapy to address the varied pathological aspects. Even if the strategy of combining drugs with different therapeutic targets is workable, the new pharmacological approach is to develop a multi-functional compound or extract to target multiple sites in the brain. Thus, the purpose of the present investigation was to demonstrate if a treatment with a standardised extract of WS can protect the human neuroblastoma cell line SK-N-SH against Aβ peptide- and acrolein-induced toxicity, decrease ROS levels and inhibit acetylcholinesterase (AChE) activity.

Materials and methods

Drugs and chemicals

A standardised extract of WS was kindly provided by Natural Remedies Private Limited, Bangalore, India. WS extract was certified to contain 2·6 % withanolides including withanosides IV (0·87 %) and V (0·65 %), withaferin A (0·56 %), withanolide A (0·20 %) and B (0·06 %), and 12-deoxy withastramonolide (0·26 %) as determined by HPLC. Dulbecco's minimum essential medium (DMEM), fetal bovine serum, penicillin/streptomycin, acrolein, acetylthiocholine iodide, neostigmine bromide, 5,5′-dithiobis-2-nitrobenzoic acid (DTNB), H2O2 and TOX-2 (XTT based) were obtained from Sigma-Aldrich Inc. The cytotoxicity detection kit based on the lactate dehydrogenase (LDH) assay was from Roche Diagnostics and 2′,7′-dichlorofluorescein diacetate (DCF-DA) was from Invitrogen. β-Amyloid peptide(25–35) was purchased from American Peptide.

25–35 preparation

The synthetic peptide Aβ25–35 was dissolved in sterilised tap water and incubated at 37°C for 72 h for fibrillisation before use as previously described( Reference Longpré, Garneau and Christen 17 ). Although both soluble and fibrillar Aβ peptide can induce oxidative stress, the latter is more efficient in causing lipid peroxidation( Reference Melo, Sousa and Garção 18 ).

Cell culture and treatment

For neuroprotective effects of WS, SK-N-SH cells, a human neuroblastoma cell line from American Type Cell Culture (ATCC), were maintained in DMEM supplemented with 10 % (v/v) fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin. SK-N-SH cells were plated at a density of 1·5 × 104 cells/well in ninety-six-well-plates (Corning) and incubated at 37°C. After 24 h of plating, the medium was completely removed and cells were kept in DMEM with antibiotics but without serum. Cells were then treated with toxic doses of Aβ (50 µg/ml) for 48 h or with acrolein (20 µm) for 24 h.

Cell viability assays

Cytotoxicity and cell survival were measured by LDH and XTT assays by using commercial kits as previously described( Reference Singh, Murthy and Ramassamy 19 ). The XTT assay measures mitochondrial dehydrogenases activity and may reflect the cellular metabolic state and serves as an indicator of cell survival. Because of the proliferative effects of the WS extract, cells may have higher dehydrogenases activity and the XTT test alone would not be sufficient to measure the neuroprotective effects. To further confirm the protective effects of the WS extract, we employed the LDH test; which measures cellular membrane integrity and is a mean of quantifying dead cells.

Intracellular reactive oxygen species level

The antioxidant activity of WS was confirmed by measuring its ability to scavenge ROS in SK-N-SH cells by using the fluorescent dye DCF-DA as previously described( Reference Singh, Murthy and Ramassamy 19 ). DCF-DA, a cell0permeable dye, is enzymically converted to a strongly fluorescent compound DCF in the presence of cellular ROS and the fluorescent intensity is directly proportional to the intracellular ROS level.

Acetylcholinesterase inhibition

The relative AChE- inhibitory activity of WS extract was determined in rat brain homogenates by a spectrophotometric method( Reference Ellman, Courtney and Andres 20 , Reference Singh and Rishi 21 ). The reaction mixture containing 50 µl of DTNB (3 mm), 25 µl WS extract, 25 µl brain homogenate from a pool of several Sprague–Dawley rats (100 µg total protein) and 100 µl of phosphate buffer. The reaction was started with the addition of 50 µl acetylthiocholine iodide (15 mm). Enzymic hydrolysis of acetylthiocholine iodide releases thiocholine, which forms a yellow 5-thio-2-nitrobenzoate anion as a result of the reaction with DTNB. The plate was read kinetically at λ 412 nm for 300 s and change in optical density/min was calculated. Neostigmine bromide (6·25 nm) was taken as a positive control.

Data analysis

All results were confirmed in four or five separate independent experiments with at least three technical replicates each time and expressed as means with their standard errors. The GraphPad InStat3 program (GraphPad Software) was used for data analysis. Statistical analysis was done by one-way ANOVA followed by Dunnett's multiple means comparison test and significance was considered when P was <0·05.

Results

Withania somnifera extract protects SK-N-SH cells against Aβ peptide- and acrolein-induced toxicity

Results in Tables 1 and 2 show that treatment with the WS extract protects SK-N-SH cells against Aβ peptide- and acrolein-induced toxicity. This protection was assessed 48 h after the addition of Aβ peptide and was significant from 1·0 and 5·0 µg/ml for the LDH and XTT assays, respectively (Table 1). Interestingly, treatment with the WS extract could also protect SK-N-SH cells against toxicity induced by acrolein from 12·5 µg/ml of WS and reached a maximum at 25 µg/ml (Table 2). To rule out any false-positive effects and interference with the XTT and LDH assays, absorbance was read and no interference was observed with the different doses of WS tested.

Table 1. Effect of treatment with Withania somnifera (WS) on SK-N-SH cell survival after 24 h with Aβ peptide (50 µg/ml) using lactate dehydrogenase (LDH) and XTT assays†

(Mean values with their standard errors of four or five separate experiments performed in triplicate at least in each group)

Mean value was significantly different from that of the Aβ group: * P < 0·05, ** P < 0·01.

† Results are expressed as percentage of control (taken as 100 %).

Table 2. Effect of treatment with Withania somnifera (WS) on SK-N-SH cell survival after 24 h with acrolein (20·0 µm) using lactate dehydrogenase (LDH) and XTT assays†

(Mean values with their standard errors of four or five separate experiments performed in triplicate at least in each group)

** Mean value was significantly different from that of the acrolein group (P < 0·01).

† Results are expressed as percentage of control (taken as 100 %).

Withania somnifera extract decreases intracellular reactive oxygen species level

Since ROS and oxidative damages are involved in the toxicity of Aβ peptide and acrolein, we investigated the antioxidant activity of WS. The WS extract was able to decrease the levels of ROS in SK-N-SH cells treated with H2O2 (500 µm), with a significant effect observed from 5·0 µg/ml (Supplementary Fig. S1).

Withania somnifera extract has potent acetylcholinesterase-inhibitory activity

The so-called cholinergic hypothesis of AD proposed that degeneration of cholinergic neurons in the basal forebrain and the associated loss of cholinergic neurotransmission in the cerebral cortex and other areas contribute significantly to the deterioration in cognitive function seen in patients with AD( Reference Francis, Palmer and Snape 22 ). AChE is the key component of cholinergic synaptic transmission and plays a major role in the termination of impulse transmission by rapid hydrolysis of the neurotransmitter acetylcholine. This serves as a rationale for the use of three Food and Drug Administration-approved AChE inhibitors in the symptomatic treatment of AD( Reference Cummings 23 ). We thus investigated the effect of WS on the activity of AChE on brain homogenates. As shown in Fig. 1, AChE activity was reduced by 30 % using 12·50 µg/ml of WS and reached about 50 % of inhibition at 50 µg/ml of WS.

Fig. 1. Effect of Withania somnifera (WS) extract on rat brain acetylcholinesterase (AChE) activity. Results are expressed as percentage of control (taken as 100 %). Neostigmine bromide (NeBr; 6·25 nm) was taken as a positive control. Values are means, with standard errors represented by vertical bars, of four or five separate experiments performed in triplicate at least in each group. ** Mean value was significantly different from that of the control group (P < 0·01).

Discussion

There is growing interest in naturally derived bioactive compounds for pharmacological application for the treatment of CCD and AD. Different strategies are being investigated such as the neuroprotective therapeutic approach based on protection against Aβ-induced neurotoxicity and oxidative damage or inhibition of AChE. In the present study, our results showed that the WS extract could significantly protect SK-N-SH cells against Aβ- and acrolein-induced toxicity, decrease ROS levels in SK-N-SH cells and inhibit the activity of AChE. The protective effect against Aβ is consistent with other studies that have suggested that WS may be neuroprotective by mediating antioxidant effects. Similar to curcuminoids and ginkgolides, withanamides from WS fruit showed potent antioxidant activity as indicated by their ability to inhibit lipid oxidation( Reference Jayaprakasam, Strasburg and Nair 24 ). In our study, the neuroprotective effect against Aβ could not be attributed to the effects of WS on ROS scavenging as its effect on ROS levels was significant from 5 µg/ml while cytoprotection was observed at lower concentrations (1 µg/ml). Other mechanisms should be investigated such as the effects of WS on redox-sensitive pathways because it has been demonstrated that withaferin A, the major constituents of the extract, could inhibit the activity of the transcription factors NF-κB, or AP-1( Reference Kaileh, Vanden Berghe and Heyerick 25 ). Also, the WS extract was more effective than withanamide A, one of the major components of WS, against Aβ because withanamide A was inactive until 12·5 µg/ml( Reference Jayaprakasam, Padmanabhan and Nair 15 ). These results indicate that other components from the WS extract are implicated in the neuroprotective effect against Aβ such as sominone, the active metabolite of withanoside IV( Reference Kuboyama, Tohda and Komatsu 14 ).

The present study demonstrated the protective effect of WS against acrolein. This result is of great interest as acrolein has been shown to be elevated in the brain from AD, from mild cognitive impairment and from preclinical AD( Reference Singh, Nam and Arseneault 26 ) and could induce neuronal and glial toxicity( Reference Thanh Nam, Arseneault and Zarkovic 9 ). Different mechanisms could underlie this protection such as the antioxidant activity of WS because acrolein has been shown to increase the production of superoxide, activate NADPH oxidase activity and deplete reduced glutathione levels( Reference Ansari, Keller and Scheff 27 ). Moreover, among the α,β-unsaturated aldehydes, acrolein reacts 110–150 times faster with glutathione than 4-hydroxy-2-nonenal or crotonal( Reference Thanh Nam, Arseneault and Zarkovic 9 ). The effects of WS on the activity of redox-sensitive pathways could also be involved as we have recently shown that the toxicity of acrolein modulates different signalling pathways( Reference Thanh Nam, Arseneault and Zarkovic 9 ).

The inhibition of AChE activity by WS from 12·5 µg/ml described here is in line with the effects of withanolides on AChE with an half maximal inhibitory concentration (IC50) ranging between 20·5 and 49·2 µg/ml( Reference Choudhary, Nawaz and ul-Haq 28 ).

WS components have been shown to induce outgrowth of axons and dendrites, and memory enhancement. Tohda & Joyashiki( Reference Tohda and Joyashiki 29 ) have demonstrated that sominone, an active metabolite of withanoside IV, could induce the phosphorylation of RET (rearranged during transfection), a receptor for glial cell line induced neurotropic factor (GDNF). Pharmacokinetics studies in mice suggested rapid oral absorption of withanolides and revealed that withaferin A has one and half times more relative bioavailability as compared with withanolide A( Reference Patil, Gautam and Mishra 30 ). Recent studies have shown that WS extract reversed brain pathology in mouse models of Alzheimer's and amyotropic lateral sclerosis, thereby indicating that certain components are bioavailable and cross the blood brain–barrier( Reference Sehgal, Gupta and Valli 31 , Reference Dutta, Patel and Rahimian 32 ).

Thus, cholinesterase-inhibiting potential along with antioxidant ability and neurotrophic and neuroprotective activity against Aβ peptide and acrolein could make WS extract and its constituents possible therapeutic agents in CCD, AD and senile dementia. Our investigation further strengthens the traditional use of WS as a nootropic agent for restoring age-related decline in mental abilities. However, further exploratory animal studies are needed to optimise therapeutic doses, to find out the right therapeutic compounds that can cross the blood–brain barrier and the duration of WS treatments aiming to yield desired beneficial outcomes; this further represents challenges as some of the components such as withaferin A are potentially cytotoxic( Reference Wadhwa, Konar and Kaul 33 ).

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/jns.2017.48

Acknowledgements

Financial support was obtained from the Natural Sciences and Engineering Research Council of Canada (NSERC) for C. R.; M. S. gratefully acknowledges financial support from the Foundation Armand-Frappier.

There were no conflicts of interest.

References

1. Schütt, T, Helboe, L, Pedersen, , et al. (2016) Dogs with cognitive dysfunction as a spontaneous model for early Alzheimer's disease: a translational study of neuropathological and inflammatory markers. J Alzheimers Dis 52, 433449.Google Scholar
2. Mattson, MP (2004) Pathways towards and away from Alzheimer's disease. Nature 430, 631639.CrossRefGoogle ScholarPubMed
3. Behl, C, Davis, JB, Lesley, R, et al. (1994) Hydrogen peroxide mediates amyloid β protein toxicity. Cell 77, 817827.CrossRefGoogle ScholarPubMed
4. Opazo, C, Huang, X, Cherny, RA, et al. (2002) Metalloenzyme-like activity of Alzheimer's disease β-amyloid. Cu-dependent catalytic conversion of dopamine, cholesterol, and biological reducing agents to neurotoxic H2O2 . J Biol Chem 277, 4030240308.Google Scholar
5. LoPachin, RM, Gavin, T, Petersen, DR, et al. (2009) Molecular mechanisms of 4 hydroxy 2-nonenal and acrolein toxicity: nucleophilic targets and adduct formation. Chem Res Toxicol 22, 14991508.Google Scholar
6. Williams, TI, Lynn, BC, Markesbery, WR, et al. (2006) Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer's disease. Neurobiol Aging 27, 10941099.Google Scholar
7. Bradley, MA, Markesbery, WR & Lovell, MA (2010) Increased levels of 4-hydroxynonenal and acrolein in the brain in preclinical Alzheimer disease. Free Radic Biol Med 48, 15701576.Google Scholar
8. Lovell, MA, Xie, C & Markesbery, WR (2001) Acrolein is increased in Alzheimer's disease brain and is toxic to primary hippocampal cultures. Neurobiol Aging 22, 187194.Google Scholar
9. Thanh Nam, D, Arseneault, M, Zarkovic, N, et al. (2010) Molecular regulations induced by acrolein in neuroblastoma SK-N-SH cell: relevance to Alzheimer's disease. J Alzheimers Dis 21, 11961216.Google Scholar
10. Chandra, V, Pandav, R, Dodge, HH, et al. (2001) Incidence of Alzheimer's disease in a rural community in India: the Indo-US study. Neurology 57, 985989.Google Scholar
11. Ven Murthy, MR, Ranjekar, PK, Ramassamy, C, et al. (2010) Scientific basis for the use of Indian Ayurvedic medicinal plants in the treatment of neurodegenerative disorders: ashwagandha . Cent Nerv Sys Agents Med Chem 10, 238246.CrossRefGoogle ScholarPubMed
12. Elsakka, M, Grigorescu, E, Stănescu, U, et al. (1990) New data referring to chemistry of Withania somnifera species. Rev Med Chir Soc Med Nat Iasi 94, 385387.Google ScholarPubMed
13. Mirjalili, MH, Moyano, E, Bonfill, M, et al. (2009) Steroidal lactones from Withania somnifera, an ancient plant for novel medicine. Molecules 14, 23732393.Google Scholar
14. Kuboyama, T, Tohda, C & Komatsu, K (2006) Withanoside IV and its active metabolite, sominone, attenuate Aβ(25–35)-induced neurodegeneration. Eur J Neurosci 23, 14171426.Google Scholar
15. Jayaprakasam, B, Padmanabhan, K & Nair, MG (2010) Withanamides in Withania somnifera fruit protect PC-12 cells from β-amyloid responsible for Alzheimer's disease. Phytother Res 24, 859863.CrossRefGoogle ScholarPubMed
16. Kumar, S, Seal, CJ, Howes, MJ, et al. (2010) In vitro protective effects of Withania somnifera (L.) dunal root extract against hydrogen peroxide and β-amyloid(1–42)-induced cytotoxicity in differentiated PC12 cells. Phytother Res 24, 15671574.Google Scholar
17. Longpré, F, Garneau, P, Christen, Y, et al. (2006) Protection by EGb 761 against β-amyloid-induced neurotoxicity: involvement of NF-κB, SIRT1, and MAPKs pathways and inhibition of amyloid fibril formation. Free Radic Biol Med 1, 8194.Google Scholar
18. Melo, JB, Sousa, C, Garção, P, et al. (2009) Galantamine protects against oxidative stress induced by amyloid-β peptide in cortical neurons. Eur J Neurosci 29, 455464.Google Scholar
19. Singh, M, Murthy, V & Ramassamy, C (2010) Modulation of hydrogen peroxide and acrolein-induced oxidative stress, mitochondrial dysfunctions and redox regulated pathways by the Bacopa monniera extract: potential implication in Alzheimer's disease. J Alzheimers Dis 21, 229247.CrossRefGoogle ScholarPubMed
20. Ellman, GL, Courtney, KD, Andres, V Jr, et al. (1961) A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 7, 8895.CrossRefGoogle ScholarPubMed
21. Singh, M & Rishi, S (2005) Plasma acetylcholinesterase as a biomarker of triazophos neurotoxicity in young and adult rats. Environ Toxicol Pharmacol 19, 471476.CrossRefGoogle ScholarPubMed
22. Francis, PT, Palmer, AM, Snape, M, et al. (1999) The cholinergic hypothesis of Alzheimer's disease: a review of progress. J Neurol Neurosurg Psychiatry 66, 137147.Google Scholar
23. Cummings, JL (2004) Alzheimer's disease. New Engl J Med 351, 5667.CrossRefGoogle ScholarPubMed
24. Jayaprakasam, B, Strasburg, GA & Nair, MG (2004) Potent lipid peroxidation inhibitors from Withania somnifera . Tetrahedron 60, 31093121.CrossRefGoogle Scholar
25. Kaileh, M, Vanden Berghe, W, Heyerick, A, et al. (2007) Withaferin A strongly elicits IκB kinase β hyperphosphorylation concomitant with potent inhibition of its kinase activity. J Biol Chem 282, 42534264.Google Scholar
26. Singh, M, Nam, DT, Arseneault, M, et al. (2010) Role of by-products of lipid oxidation in Alzheimer's disease brain: a focus on acrolein. J Alzheimers Dis 21, 741756.Google Scholar
27. Ansari, MA, Keller, JN & Scheff, SW (2008) Protective effect of Pycnogenol in human neuroblastoma SH-SY5Y cells following acrolein-induced cytotoxicity. Free Radic Biol Med 45, 15101519.CrossRefGoogle ScholarPubMed
28. Choudhary, MI, Nawaz, SA, ul-Haq, Z, et al. (2005) Withanolides, a new class of natural cholinesterase inhibitors with calcium antagonistic properties. Biochem Biophys Res Commun 334, 276287.Google Scholar
29. Tohda, C & Joyashiki, E (2009) Sominone enhances neurite outgrowth and spatial memory mediated by the neurotrophic factor receptor, RET. Br J Pharmacol 157, 14271440.CrossRefGoogle ScholarPubMed
30. Patil, D, Gautam, M, Mishra, S, et al. (2013) Determination of withaferin A and withanolide A in mice plasma using high-performance liquid chromatography-tandem mass spectrometry: application to pharmacokinetics after oral administration of Withania somnifera aqueous extract. J Pharm Biomed Anal 80, 203212.Google Scholar
31. Sehgal, N, Gupta, A, Valli, RK, et al. (2012) Withania somnifera reverses Alzheimer's disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci U S A 109, 35103515.Google Scholar
32. Dutta, K, Patel, P, Rahimian, R, et al. (2017) Withania somnifera reverses transactive response DNA binding protein 43 proteinopathy in a mouse model of amyotrophic lateral sclerosis/frontotemporal lobar degeneration. Neurotherapeutics 14, 447462.Google Scholar
33. Wadhwa, R, Konar, A & Kaul, SC (2016) Nootropic potential of ashwagandha leaves: beyond traditional root extracts. Neurochem Int 95, 109118.Google Scholar
Figure 0

Table 1. Effect of treatment with Withania somnifera (WS) on SK-N-SH cell survival after 24 h with Aβ peptide (50 µg/ml) using lactate dehydrogenase (LDH) and XTT assays†(Mean values with their standard errors of four or five separate experiments performed in triplicate at least in each group)

Figure 1

Table 2. Effect of treatment with Withania somnifera (WS) on SK-N-SH cell survival after 24 h with acrolein (20·0 µm) using lactate dehydrogenase (LDH) and XTT assays†(Mean values with their standard errors of four or five separate experiments performed in triplicate at least in each group)

Figure 2

Fig. 1. Effect of Withania somnifera (WS) extract on rat brain acetylcholinesterase (AChE) activity. Results are expressed as percentage of control (taken as 100 %). Neostigmine bromide (NeBr; 6·25 nm) was taken as a positive control. Values are means, with standard errors represented by vertical bars, of four or five separate experiments performed in triplicate at least in each group. ** Mean value was significantly different from that of the control group (P < 0·01).

Supplementary material: File

Singh and Ramassamy supplementary material

Figure S1

Download Singh and Ramassamy supplementary material(File)
File 117.8 KB