Open access peer-reviewed chapter

Bioactive Molecules from Indian Medicinal Plants as Possible Candidates for the Management of Neurodegenerative Disorders

Written By

Uma Ranjan Lal and Snigdha Lal

Submitted: 18 October 2019 Reviewed: 08 March 2020 Published: 07 May 2020

DOI: 10.5772/intechopen.92043

From the Edited Volume

Bioactive Compounds in Nutraceutical and Functional Food for Good Human Health

Edited by Kavita Sharma, Kanchan Mishra, Kula Kamal Senapati and Corina Danciu

Chapter metrics overview

829 Chapter Downloads

View Full Metrics

Abstract

The present review gives an account of various bioactive molecules obtained from Indian medicinal plants for neurological degenerative disorders. Emphasis is laid on their correlation with the plants used in traditional system of medicine in India. The methodology involved in present review was enlisting of medicinal plants used for neurodegenerative disorders followed by their chemistry. A correlation with the chemical constituents and their recent findings has been done. Many medicinal plants such as Aloe vera and Bacopa monnieri have documented correlations and also need to be explored more. Molecules like garcinol (34), which was originally an anticancer compound, have good correlation as neuroprotective agent. Likewise many plants that have not been explored but are used in traditional system of medicine have also been listed. Jaggery and honey, which are used in traditional formulations in large quantity, also have natural products that are used as neuroprotective agents. In conclusion, a lot more study is required to correlate the medicinal plants and herbal formulations to have much more natural products for neurodegenerative disorders.

Keywords

  • ayurvedic formulations
  • Indian medicinal plants
  • aloin
  • garcinol

1. Introduction

Neurodegeneration is defined as a complex process that involves progressive damage to the brain with neuronal loss leading to incurable and devitalizing consequences. The neurons, which get degenerated, are not restored, which result in impaired cognition and neurological disorders leading to depression, schizophrenia, Alzheimer’s disease, dementia, epilepsy, cerebral ischemia, and parkinsonism [1, 2]. These diseases involve various characteristic pathological and molecular features affecting neurons in various regions of the brain. Neurodegenerative diseases are accelerated by the way we live our daily lives. As per recent report by Indian Council of Medical Research (ICMR), the proportion of deaths estimated due to lifestyle diseases has increased from 37.09% in 1990 to 61.8% in 2016 [3]. Thus, there is an immense need for the development of strategies for its prevention as a number of patients suffering from lifestyle diseases are increasing day by day. Ayurveda is considered an old age traditional medicine of Indian practice involving considerable usage of plants and herbal preparations, which are known to cure various disorders. Various classical formulations for neurodegenerative diseases are listed in Ayurvedic Formulary of India (AFI), which also provides a file of information regarding single drugs of plant, animal, and mineral origin, providing their official names and English equivalents for their easy identification [4]. In present review, the major plants listed in the formulations indicated as neuroprotective is discussed. Present review outlines the chemical constituents and pharmacology of the listed plants. The sole aim of present review is to extract the hidden potential ancient formulations supported by modern findings so as to enhance their acceptability in masses. Further, a systematic approach for their chemical standardization is a need as it will be supportive in identifying therapeutic molecules, stating the need for combining scientific interpretations and traditional knowledge.

Advertisement

2. Indian medicinal plants and neurodegenerative diseases

The present sections describe plant material along with their chemical constituents and related pharmacology pertaining to neurodegenerative disorders. Few molecules with prominent activity have also been discussed.

Advertisement

3. Aloe vera (L.) Burm.f.

A. vera is a succulent, evergreen, perennial herb, which is being cultivated all over the world mainly for the purpose of agricultural and medicinal uses. Anthraquinones (aloe-emodin (1), chrysophanol (2), and a bitter reddish-yellow exudate containing majorly anthrones (aloin A and B (3)), aloe-emodin (4) [5], and few aloe resins such as aloeresin (5) and aloenin (6)) [6] are the major constituents in aloe (Figure 1). Aloe-emodin 4 is reported to enhance cognition against scopolamine-induced cognitive impairment in mice [7]. Aloin 3 is shown to decrease intracellular ROS generation and reduced Ca2+ production, which is responsible for depolarization and death of the neuron, suggesting it as a useful and alternative therapy for cerebrovascular diseases [8]. A. vera gel demonstrated antioxidant and anti-inflammatory activities when given to the rats with sciatic nerve reperfusion injury [9].

Figure 1.

Structures of compounds present in A. vera.

Advertisement

4. Terminalia chebula Retz

Terminalia chebula (Combretaceae) commonly called Haritaki, is used in ayurvedic formulations, also reported to demonstrate neuroprotective activities. The principle constituents in pericarp of T. chebula fruits are phenolics (chebulic acid (7), chebulagic acid (8), chebulinic acid (9), ellagic acid (10), gallic acid (11)) [10] and triterpenoid glycosides chebuloside II (12) (Figure 2) [11]. Pericarp of fruits of T. chebula constitutes Triphala (one of the popular formulations of ayurvedic system of medicine). The methanolic (70%) extract of the fruit Fructus chebulae has been shown to rescue cerebral ischemia by protecting neurons from degeneration. The in-vitro study and in-vivo studies have shown promising results [12]. Also, there is a promising decrease in the levels of malondialdehyde (MDA), NO, and microglial death stimulated by lipopolysaccharide (LPS) in the cells after treating with the extract [13]. The underlying mechanism might be the inhibition of inflammatory and oxidative processes. T. chebula constituents such as chebulagic acid, chebulinic acid, and ellagic acid have shown to be neuroprotective on various cell lines by showing its effect on various targets [14, 15, 16].

Figure 2.

Structures of compounds present in T. chebula.

Advertisement

5. Honey and Jaggery

Both Honey and Jaggery are used in making of many ayurvedic preparations, and they constitute major part of many formulations [4]. Honey, a sweet and viscous substance, is produced by the honey bees (Apis spp.). It contains sugar and others such as minerals, proteins, essential oils, and flavonoids. Dietary polyphenols found in honey as well as other food and plant materials can prevent neurodegenerative disease in various ways [17, 18, 19, 20, 21, 22]. These include oxidative protection of neurons [17], enhancement of neuronal function and regeneration [18], protection of neurons from Aβ-induced neuronal injury and neurotoxicity [19], protection of hippocampal cells against nitric oxide-induced toxicity [20], and modulation of neuronal and glial cell signaling pathways [21]. Luteolin (13) shows neuroprotective activity via prevention of microglia-associated inflammation in the hippocampus of aged rats [22]. Other flavonoids such as quercetin (14) and kaempferol (1) have also shown to be neuroprotective in various models. Kaempferol’s (15) neuroprotective effect was confirmed by the histochemical findings where it prevented the loss of TH-positive neurons induced by MPTP [23, 24, 25]. Luteolin 13, a flavone, has a significant role in treating CNS disorders through various mechanisms [26]. The activity of luteolin 13was also reported at its microglial transcriptome level for the treatment of CNS disorders via anti-inflammatory activity [27]. Pretreatment of mice with luteolin 13 was expected to reduce the frequency of seizure in the PTZ mice model [28]. Luteolin 13 also has the potential to improve the activity of oxygen-glucose deprivation/reperfusion (OGD/R) induced neurons in a dose-dependent manner by enhancing the Na+/K+-ATPase activity suggesting a potential molecule to treat cerebral ischemia [29]. Apigenin (16), a trihydroxyflavone, was reported to inhibit the release of glutamate from hippocampal nerve terminals, which might be useful in treating epilepsy [30]. The role of apigenin (16) in the treatment of cognition was demonstrated by attenuating the Aβ-induced cytotoxicity in rat cortical neurons but having no intervention with oxidative stress. The biological activities of the flavonoids might be due to hydroxyl groups at R2 and R3 position influencing various cellular events eventually leading to apoptosis [27, 28, 29, 30]. Other phenolics such as ferulic acid (17) exert neuroprotective effect through middle cerebral artery occlusion [31] and by decreasing the number of microglia/macrophages after cerebral ischemia/reperfusion injury in rats [32]. Chlorogenic acid (18) present in honey exerts a neuroprotective effect against methyl mercury-induced apoptosis in pheochromocytoma-12 (PC12) cell lines. It prevents the generation of reactive-oxygen species (ROS), suppressing the decreasing action of glutathione peroxidase (GPx) and GSH and attenuating apoptosis by the activation of caspase-3 [33] and also inhibits the activity of acetylcholine esterase and MDA in the hippocampus as well as in the frontal cortex in mice [34]. Honey as such has also shown to be neuroprotective in male Sprague-Dawley rats, and there was a decrease in the thiobarbituric acid reactive substance levels in the rats, which were pretreated with honey [35]. Chrysin (19) improved the morphological integrity of nigrostriatal neurons and increased the endogenous levels of BDNF, S100B, NGF, and GDNF in mice striatum improving behavioral and better muscular coordination [36]. Its lipophilic nature and bioflavonoid nucleus confers an added advantage to grow as a compelling therapeutic agent for neurological disorders. Pinocembrin (20) elevated glutamate level, which was an important excitatory neurotransmitter produced by global cerebral I/R [37].

Jaggery is used in most of the ayurvedic formulations [4]. Traditionally, it is indicated for treating neurodegenerative disorders. The chemistry of sugar cane reveals that it is rich in phenolics and their glycosides [38], flavonoid glycosides, and flavones [39]. Tricin-7-glucoside (21) was reported to possess neuroprotective activity against cerebral ischemia via reduced expression of NF-κB and HMGB1. SH-SY5Y cells, which were pretreated with tricin (21a), reduced the apoptosis induced by OGD. Attenuation of histopathological damage reduced brain edema was reported in animal models of ischemia along with reduced NF-κB and HMGB1 expression [40]. It also has potential for Alzheimer’s disease [41] (Figure 3).

Figure 3.

Structures of compounds present in Jaggery and honey.

Advertisement

6. Bacopa monnieri L.

Bacopa monnieri L. (Scrophulariaceae) is notable ayurvedic medicinal plant and referred to have impact on brain function and is a neural tonic to improve intelligence and cognition [42]. Normal formulation Illumina® containing B. monnieri (leaf dry extract 43%, containing bacoside, 20%) decreased of 8-iso-PFG2α and ROS/RNS generation in the rat brain, consequently, decreasing the basal and H2O2 and amyloid β peptide-incited oxidative stress [43]. In another study, standardized extract of B. monnieri (Bacoside A (22) content 82 ± 0.5%) when given in the doses of 5 and 10 mg/kg, orally, shows a dose-related increment in superoxide dismutase, catalase, glutathione peroxidase activities in the frontal cortex, striatum, and hippocampus [44]. B. monnieri, when administered orally, improves cognitive impairment and neurodegeneration in rats. Immunohistological recognition of superoxide dismutase (Cu/Zn-SOD) and histopathological changes in the CA1 region of the hippocampus were observed [45]. An extract containing 5% (w/w) of the saponins, which contains bacoside A3, bacopasaponin X, bacopasaponin C (23), bacopaside II (24), and bacopaside I (25), shows improvements in cognitive abilities and neuroprotective impacts in Alzheimer’s disease model [46, 47]. When rats treated with bacoside A (22), huge changes were found in the degrees of both nonenzymatic and enzymatic antioxidants, which recommends that bacoside A 22 improves B. monnieri antioxidant status in rat brain [48]. Other saponins such as (26-28) reported from this plant have been shown in Figure 4. A survey of 10 years of research at Swinburne University done by Con Stough et al. recommends that an extract of B. monnieri (CDRI 08: KeenMind) is a safe and effectual cognitive enhancer [49]. B. monnieri extract suppressed the generation of free radical levels and indicated critical assurance against 3-nitropropionic acid (3-NPA)-mediated cytotoxicity in dopaminergic (N27 cell lines) [50]. B. monnieri standard extract containing 55.34% of bacosides indicated a defensive impact on ischemia-induced memory hindrance and diminished the infarct size in the ischemic brain. It additionally demonstrated a critical increment in catalase action and exhaustion in lipid peroxidation, nitrite, and nitrate activity [51]. Neuronal cell cultures when treated with B. monnieri extract shielded neurons from β-amyloid-induced cell toxicity. The extract gave protection to cell cultures against glutamate-induced excitotoxicity since it was not able to repress glutamate-mediated toxicity [52]. In Alzheimer’s disease animal model (C57/Bl6 mice), B. monnieri extract has decreased lipoxygenase action and hydrogen peroxide-induced lipid peroxidation. B. monnieri has neuroprotective mechanisms, and it lessens β-amyloid deposits in the brain of C57/Bl6 mice Alzheimer’s disease animal model [53]. B. monnieri extract have also shown to have protective effect against rotenone induced Parkinson’s disease in PC-12 cell lines [54].

Figure 4.

Structures of compounds present in B. monnieri.

Advertisement

7. Asparagus racemosus Willd.

Methanolic extract of Asparagus racemosus decreased the degrees of cytokines, malondialdehyde (as a marker of lipid peroxides formation), and nitric oxide with a critical increment in the levels of catalase, superoxide dismutase, and glutathione, concluding its neuroprotective activity [55]. Its root extract (100 mg/kg) cured region-specific neurodegeneration created in Swiss albino mice [56] and also demonstrated dose-dependent improvement in memory after histochemical and behavioral studies. A noteworthy decrease in transfer latency time and a significant increment in acetylcholinesterase (AchE) staining in histochemical identification were observed suggesting antioxidant, cholinergic, and neuroprotective properties of A. racemosus [57]. EuMil, a formulation containing standardized extracts A. racemosus Willd., has been used for stress-related problems and has been found to re-establish modified degree of nor-adrenaline, 5-hydroxytryptamine, and dopamine normal (100 mg/kg, p.o. 14 days) [58]. Withania somnifera and A. racemosus together have shown noteworthy impacts in cell viability test, lactic dehydrogenase, malondialdehyde, glutathione disulfide, glutathione, nerve growth factor, pro-brain-derived growth factor levels, and reactive oxygen species generation [59]. Ovariectomized adult female Wistar rats showed noteworthy upregulation of estrogen receptors (ERα and ERβ) in hippocampus and frontal cortex area alongside enhancement in the levels of brain-derived neurotrophic factor. Upregulation of estrogen receptors and enhancement in the levels of brain-derived neurotrophic factor can be filled in as proof for neuroprotective impact of ethanolic concentrate of A. racemosus roots [60, 61]. Significant protection is seen after the supplementation of Mentat (BR-16A) is an herbal psychotropic preparation, containing A. racemosus against ethanol withdrawal-induced decrease of pentylenetetrazole threshold in rats and mice [62]. Sarsasapogenin, a steroidal saponin from A. racemosus, has been studied for neuroprotective impact in Alzheimer’s disease. Sarsasapogenin (29) indicated noteworthy restraint of butyrylcholinesterase, monoamine oxidase-B, beta-secretase 1, and acetylcholinesterase, key enzymes related to the pathogenesis of Alzheimer’s disease. At the point when tested against Aβ42 and H2O2-interceded cytotoxicity, sarsasapogenin showed a huge neuroprotective impact on PC12 cells. These discoveries recommended that sarsasapogenin can go about as a multi-target directed ligand and as a reasonable lead compound for treating different elements engaged with the pathogenesis of Alzheimer’s disease [63]. Alterations in the normal levels of neurotransmitters, glutamate, acetylcholinesterase, dopamine, and protein because of worldwide cerebral ischemia were normalized by standardized A. racemosus Willd. root methanolic extract, appeared by the abatement in the degree of glutamate, acetylcholinesterase, and increment in dopamine levels and protein levels. Group treated with methanolic root extract of A. racemosus Willd. shown 85% neuronal protection in the CA1 area of the hippocampus. This demonstrated the cerebroprotective role of A. racemosus Willd. [64].

Advertisement

8. Foeniculum vulgare Mill.

Foeniculum vulgare extract reduced the amnesic effect and memory deficits in mice induced due to aging. F. vulgare extract demonstrated inhibition of acetylcholine, and in the exteroceptive behavioral model, it increased the step-down latency in mice significantly [65]. Fennel essential oil inhalation inhibits beta-amyloid (1-42)-induced depression and anxiety and also indicates that it may have further clinical applications [66]. There were improvements in Parkinson’s disease in the animal model produced by F. vulgare Mill. essential oil [67]. Clinically fennel supplementation to obese middle-aged women decreased bodyweight, reduction in serum Aβ protein along with improvements in cognitive functions and metabolic profiling [68]. It normalized the expression levels of oxidative stress markers [Superoxide dismutase and Peroxiredoxin-6 (Prdx6)] and APP isoforms (APP common, 770 and 695) and also improved the Pb-induced morphological deterioration of cortical neurons [69].

Advertisement

9. Azadirachta indica A. Juss.

Commonly known as Neem in Indian subcontinent, Azadirachta indica has been shown to attenuate cisplatin-induced neurotoxicity in rats, and it also had neuroprotective effect on cerebral post-ischemic reperfusion and hypoperfusion [70, 71]. A. indica extracts have shown to be anti-oxidative and anti-apoptotic neuroprotective in Parkinson-induced functional damage [72]. A. indica standardized leaf extract (total bitters 4.3%) has shown to be neuroprotective in partial sciatic nerve injury in rats as evidenced from anti-inflammatory, antioxidant, and anti-apoptotic studies [73].

Advertisement

10. Picrorhiza kurroa Royle ex Benth

Picrorhiza kurroa, an ayurvedic herb, has been shown to potentiate photochemotherapy in vitiligo [74]. Apocynin (30) from P. kurroa has shown to be neuroprotective in vivo [75]. It also shows protective effect in a mouse model of chemically induced colitis [76]. It (4-hydroxy-3-methoxy-acetophenone, 30) mediates long-lasting memory recovery, helps in hippocampal neuroprotection, and reduces glial cell activation after transient global cerebral ischemia in rats [77]. Interestingly, the concentration of picrosides I (31) and II (32) and apocynin 30 (iridoid rich fraction) in plasma (C max) was found to be 244.9, 104.6, and 502 ng/ml with half-life (t 1/2) 14, 8, and 6 h, respectively [78]. P. kurroa also prevents memory deficits by inhibiting NLRP3 Inflammasome Activation and BACE1 Expression in 5xFAD Mice [79]. Acylated iridoid glycosides with hyaluronidase inhibitory activity from the rhizomes of P. kurroa Royle ex Benth have recently been isolated [80]. Therapeutic potentials of plant iridoids in Alzheimer’s and Parkinson’s diseases have separately been reviewed recently [81].

11. Berberine from Berberis aristata DC

Berberine (33) has been shown to be neuroprotective through the Nrf2 upregulation and also alleviates rotenone-induced cytotoxicity by antioxidation and activation of PI3K/Akt signaling pathway in SH-SY5Y cells [82]. Neuroprotective effects of berberine have also been confirmed in animal models of Alzheimer’s disease [83]. Berberine nanoparticles have shown protective effect against LPS-induced neurodegenerative changes [84]. Berberine confers neuroprotection in coping with focal cerebral ischemia by targeting inflammatory cytokines [85]. Berberine had also shown protective effect against the altered intrinsic properties of the CA1 neurons induced by Aβ neurotoxicity [86]. There are many pathways by which berberine acts to protect neurons and has recently been reviewed. Authors have concluded that it being a potential candidate for combating neurodegenerative diseases [87, 88].

12. Garcinol from Garcinia indica Choisy

Garcinol (34) is one of the major constituents of Garcinia indica (a plant found in the region of Western Ghats of India). Garcinol can effectively restore the balance between the neurotransmitters glutamate and the γ-aminobutyric acid (GABA), rescue neural precursor cells, and promote their rapid growth. It regulates the expressions of glutamic acid decarboxylase 65 and GABAA receptors, preventing hyperactivation of NMDA receptor and the resultant excitotoxicity. It enhances memory and cognition in C57BL/6 mice, significantly lowering epileptic seizure scores [89, 90]. It also serves as a strong inhibitor of histone acetyltransferases (HAT), thus contributing protection against rapid neurodegeneration in parkinsonian brain [91, 92]. Garcinol helps in restoration of dopamine potency and has homocysteine lowering ability as well as have been shown to counter LDOPA-induced dyskinesia in PD model, demonstrate its worthiness as a potential drug candidate against Parkinson’s Disease [93, 94]. Garcinol also exhibits desirable anti-cholinesterase properties by inhibiting the enzyme acetyl cholinesterase with an IC50 value of 0.66 μM, and it improves the neuronal count in hippocampal regions following administration of pentylenetetrazole (PTZ). In cultured rat cortical progenitor cells, garcinol can reduce cell death associated with growth factor deprivation. It also promotes neurite outgrowth in epidermal growth factor-responsive neural precursor cells and supports the survival of neurons [90]. In unilaterally 6-hydroxydopamine (6-OHDA)-lesioned hemi parkinsonian mice, 5 mg/kg of garcinol co-treatment with L-DOPA effectively controlled the axial, limb, and orofacial (ALO) score for dyskinesia analysis. Following the administration of garcinol, a decreased expression of c-Fos, FRA-2, and ARC genes has been visualized [94], which is usually over-activated in L-DOPA induced dyskinesia [95]. Moreover, methanolic extract of G. indica, which is known to contain garcinol, effectively elevates dopamine level in the striatum and confers neuroprotection to dopaminergic neurons in 6-OHDA lesioned experimental rats [96]. Even pharmacological inhibition of HATs by garcinol can notably suppress MPP+-induced cell death due to reduction in ATP content [97]. In silico studies on the molecular interaction between garcinol and the active sites of COMT and MAO-B revealed that garcinol can potentially inhibit the activity of the two enzymes, similar to their known inhibitors [93]. In conclusion, garcinol may prove to be a dependable remedial measure in PD therapeutics since its inhibition of MAO-B and COMT can be correlated to increase the availability of dopamine as well as prevent the generation of toxic dopamine metabolites including homocysteine, 3-omethyldopa, 3-methoxytyramine, and 3,4-dihydroxyphenylacetaldehyde [98] (Figure 5).

Figure 5.

Structures of compounds present in various plants for neuroprotective activity.

13. Nardostachys jatamansi (D. Don)DC.(Jatamansi)

The plant Nardostachys jatamansi is well exploited in Ayurvedic system of medicine for its role in neurological disorders. Various phytochemicals have been reported from this plant by various authors. Its sedative and antidepressant activity, its mechanism (inhibition of MAO and GABA), and its role in rat cerebral ischemia have been documented [99]. Anticonvulsant activity and neurotoxicity profile of the plant have been generated [100]. The extracts of N. jatamansi have been found to attenuate 6-hydroxydopamine-induced parkinsonism in rats, as proven by behavioral, neurochemical, and immunohistochemical studies [101]. It also improves learning and memory in mice. It also has stress modulating antioxidant effect. Its formulation was also found to enhance the learning and memory process in rats [102, 103, 104]. It has shown neuroprotective efficacy in conjunction with selenium in cognitive impairment [105]. N. jatamansi root extract was found to modulate the growth of IMR-32 and SK-N-MC neuroblastoma cell lines through MYCN-mediated regulation of MDM2 and p53 [106]. Novel Sesquiterpeniods and Anti-neuroinflammatory metabolites from N. jatamansi have also been isolated [107]. Compounds such as Desoxo-narchinol A 35 and Narchinol B 36 isolated from N. jatamansi have been shown to exert anti-neuroinflammatory effects by upregulating nuclear transcription factor erythroid-2-related factor 2/heme oxygenase-1 signaling [108].

14. Other plants

Many plants, which have been used in traditional formulations for neurological disorders, need to be phytochemically explored and correlated with the modern findings. Table 1 gives the list of those plants with reported neuroprotective activity.

Plant name Part used Reports on neuroprotective activity Reference
Fumaria indica (Hausskn.) Pugsley Leaf Significant activity of ethanolic extract on rat cognitive dysfunctions. Potential antianxiety activity of leaf extract; preclinical study [109, 110]
Alhagi pseudalhagi (M. Bieb) Desv. ex B. Keller & Shap. Whole plant Traditionally used for neuroprotective disorders. Compounds having neuroprotective activity like flavanone glycosides and alkaloids like β-phenethylamine and tetrahydroisoquinoline have been reported [111, 112, 113, 114]
Pluchea lanceolata (Oliver & Hiern.) Leaf Protection of hippocampal neurons from endothelin-1 induced ischemic injury to ameliorate cognitive deficits; protective effect against aluminum chloride-induced neurotoxicity in Swiss Albino mice; protective effect on LPS-induced neuro-inflammation in C6 rat glial cells [115, 116, 117]
Premna mucronata Roxb. Whole plant Luteolin and apigenin are reported, and they are reported to be neuroprotective [118]
Semecarpus anacardium L.f. Fruits Stress induced neuroprotective activity [119]
Sida cordifolia L. Whole plant Ameliorative effect in parkinsonism [120]
Tinospora cordifolia (Thunb.) Miers. Stems Suppresses neuro-inflammation in Parkinsonian Mouse Model; potential neuro-regenerative candidate against glutamate induced excitotoxicity: an in vitro perspective [121, 122, 123]
Trichosanthes dioica Roxb. Rhizome Neuropharmacological properties of root [124]
Strobilianthes ciliatus (Nees.) No such reports

Table 1.

List of plants whose exploration is required, as they are used tremendously in traditional system of medicine in India.

15. Conclusions

In present review we focus on evidence to prevent neurodegenerative disorders in various studies (in vitro and in vivo). The mentioned medicinal plants play their protective roles via increased SOD and catalase levels, restoration of GSH, and decreased MDA levels and also protect neurons against ROS as antioxidant activities. The neuroprotective effects of the mentioned plants occur via reduction of inflammatory cytokines as well as enhancement of anti-inflammatory cytokines, inhibition of the acetylcholinesterase activity, and decreased MDA levels in the neural system via modulating GABAergic and glutamatergic neurons and also increasing amount of amino acids and serotonin (5-HT) in the neurotransmitters systems. Based on the evidence produced, it is suggested that more exploration of traditional formulations is required, and also repurposing in natural products is also required to a greater extent, as is evident from recent reports on garcinol and berberine.

Conflict of interest

Authors do not have any conflict of interest.

References

  1. 1. Mattson MP. Metal-catalyzed disruption of membrane protein and lipid signaling in the pathogenesis of neurodegenerative disorders. Annals of the New York Academy of Sciences. 2004;1012:37-50
  2. 2. Saxena S, Caroni P. Selective neuronal vulnerability in neurodegenerative diseases: From stressor thresholds to degeneration. Neuron. 2011;71:35-48
  3. 3. Available from: https://pib.gov.in/Pressreleaseshare [Accessed: 31 January 2020]
  4. 4. Anonymous. Ayurvedic Formulary of India, (part I, II, III). Government of India, Ministry of Health and Family Welfare Department of Indian Systems of Medicine and Homoeopathy; 2011
  5. 5. Saleem R, Faizi S, Deeba F, Siddiqui BS, Qazi MH. Anthrones from Aloe barbadensis. Phytochemistry. 1997;45:1279-1282. DOI: 10.1016/S0031-9422(97)00107-6
  6. 6. Speranza G, Gramatica P, Dadá G, Manitto P. Aloeresin c, a bitter C,O-diglucoside from cape aloe. Phytochemistry. 1985;24:1571-1573. DOI: 10.1016/S0031-9422(00)81068-7
  7. 7. Tao L, Xie J, Wang Y, Wang S, Wu S, Wang Q , et al. Protective effects of aloe-emodin on scopolamine-induced memory impairment in mice and H2O2-induced cytotoxicity in PC12 cells. Bioorganic & Medicinal Chemistry Letters. 2014;24:5385-5389. DOI: 10.1016/j.bmcl.2014.10.049
  8. 8. Chang R, Zhou R, Qi X, Wang J, Wu F, Yang W, et al. Protective effects of aloin on oxygen and glucose deprivation-induced injury in PC12 cells. Brain Research Bulletin. 2016;(121):75-83. DOI: 10.1016/j.brainresbull.2016.01.001
  9. 9. Guven M, Gölge UH, Aslan E, Sehitoglu MH, Aras AB, Akman T, et al. The effect of Aloe vera on ischemia-reperfusion injury of sciatic nerve in rats. Biomedicine & Pharmacotherapy. 2016;79:201-207. DOI: 10.1016/j.biopha.2016.02.023
  10. 10. Lee DY, Kim HW, Yang H, Sung SH. Hydrolyzable tannins from the fruits of Terminalia chebula Retz and their α-glucosidase inhibitory activities. Phytochemistry. 2017;137:109-116. DOI: 10.1016/j.phytochem.2017.02.006
  11. 11. Kundu AP, Mahato SB. Triterpenoids and their glycosides from Terminalia chebula. Phytochemistry. 1993;32:999-1002. DOI: 10.1016/0031-9422(93)85243-K
  12. 12. Gaire BP, Kim H. Neuroprotective effects of Fructus Chebulae extracts on experimental models of cerebral ischemia. Journal of Traditional Chinese Medicine. 2014;34:69-75. DOI: 10.1016/s0254-6272(14)60057-1
  13. 13. Gaire BP, Jamarkattel-Pandit N, Lee D, Song J, Kim JY, Park J, et al. Terminalia chebula extract protects OGD-R induced PC12 cell death and inhibits LPS induced microglia activation. Molecules. 2013:3529-3542. DOI: 10.3390/molecules18033529
  14. 14. Kim HJ, Kim J, Kang KS, Lee KT, Yang HO. Neuroprotective effect of chebulagic acid via autophagy induction in SH-SY5Y cells. Biomolecules & Therapeutics. 2014;22:275-281. DOI: 10.4062/biomolther.2014.068
  15. 15. Shen YC, Juan CW, Lin CS, Chen CC, Chang CL. Neuroprotective effect of Terminalia chebula extracts and ellagic acid in Pc12 cells. African Journal of Traditional, Complementary and Alternative Medicines (AJTCAM). 2017;14:22-30. DOI: 10.21010/ajtcam.v14i4.3
  16. 16. Song JH, Shin MS, Hwang GS, Oh ST, Hwang JJ, Kang KS. Chebulinic acid attenuates glutamate-induced HT22 cell death by inhibiting oxidative stress, calcium influx and MAPKs phosphorylation. Bioorganic & Medicinal Chemistry Letters. 2018;28:249-253. DOI: 10.1016/j.bmcl.2017.12.062
  17. 17. Cirmi S, Ferlazzo N, Lombardo GE, Ventura-Spagnolo E, Gangemi S, Calapai G, et al. Neurodegenerative diseases: Might Citrus flavonoids play a protective role? Molecules. 2016;21:1312
  18. 18. Inanami O, Watanabe Y, Syuto B, Nakano M, Tsuji M, Kuwabara M. Oral administration of (−) catechin protects against ischemia-reperfusion-induced neuronal death in the gerbil. Free Radical Research. 1998;29:359-365
  19. 19. Youdim KA, Joseph JA. A possible emerging role of phytochemicals in improving age-related neurological dysfunctions: A multiplicity of effects. Free Radical Biology & Medicine. 2001;30:583-594
  20. 20. Vauzour D, Vafeiadou K, Rice-Evans C, Williams RJ, Spencer JP. Activation of pro-survival Akt and ERK1/2 signalling pathways underlie the anti-apoptotic effects of flavanones in cortical neurons. Journal of Neurochemistry. 2007;103:1355-1367
  21. 21. Jang S, Dilger RN, Johnson RW. Luteolin inhibits microglia and alters hippocampal-dependent spatial working memory in aged mice. The Journal of Nutrition. 2010;140:1892-1898
  22. 22. Xu B, Li X-X, He G-R, Hu J-J, Mu X, Tian S, et al. Luteolin promotes long-term potentiation and improves cognitive functions in chronic cerebral hypoperfused rats. European Journal of Pharmacology. 2010;627:99-105
  23. 23. Hu P, Wang M, Chen W-H, Liu J, Chen L, Yin S-T, et al. Quercetin relieves chronic lead exposure-induced impairment of synaptic plasticity in rat dentate gyrus in vivo. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2008;378:43-51
  24. 24. Kumar A, Sehgal N, Kumar P, Padi S, Naidu P. Protective effect of quercetin against ICV colchicine-induced cognitive dysfunctions and oxidative damage in rats. Phytotherapy Research. 2008;22:1563-1569
  25. 25. Li S, Pu X-P. Neuroprotective effect of kaempferol against a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridineinduced mouse model of Parkinson’s disease. Biological & Pharmaceutical Bulletin. 2011;34:1291-1296
  26. 26. Ashaari Z, Hadjzadeh MAR, Hassanzadeh G, Alizamir T, Yousefi B, Keshavarzi Z, et al. The flavone luteolin improves central nervous system disorders by different mechanisms: A review. Journal of Molecular Neuroscience. 2018;65:491-506. DOI: 10.1007/s12031-018-1094-2
  27. 27. Dirscherl K, Karlstetter M, Ebert S, Kraus D, Hlawatsch J, Walczak Y, et al. Correction: Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype. Journal of Neuroinflammation. 2010;7:1-2. DOI: 10.1186/1742-2094-7-3
  28. 28. Hosseinzadeh H, Nassiri-Asl M. Neuroprotective effects of flavonoids in epilepsy. In: Brahmchari G, editor. Neuroprotective Natural Products Clinical Aspects Mode of Action, Chapter 10. Weinheim, Germany : Wiley-VCH; 2017. pp. 279-291. ISBN: 978-3-527-34186-3
  29. 29. Fang L, Zhang M, Ding Y, Fang Y, Yao C, Zhang X. Protective effects of luteolin on neurons against oxygen-glucose deprivation/reperfusion injury via improving Na+/K+-ATPase activity. Zhongguo Zhongyao Zazhi. 2010;35:1051-1054. DOI: 10.4268/cjcmm20100825
  30. 30. Kang SS, Lee JY, Choi YK, Kim GS, Han BH. Neuroprotective effects of flavones on hydrogen peroxide-induced apoptosis in SH-SY5Y neuroblostoma cells. Bioorganic & Medicinal Chemistry Letters. 2004;14:2261-2264. DOI: 10.1016/j.bmcl.2004.02.003
  31. 31. Koh P-O. Ferulic acid prevents the cerebral ischemic injury-induced decrease of Akt and bad phosphorylation. Neuroscience Letters. 2012;507:156-160
  32. 32. Cheng C-Y, Su S-Y, Tang N-Y, Ho T-Y, Chiang S-Y, Hsieh C-L. Ferulic acid provides neuroprotection against oxidative stress-related apoptosis after cerebral ischemia/reperfusion injury by inhibiting ICAM-1 mRNA expression in rats. Brain Research. 2008;1209:136-150
  33. 33. Li Y, Shi W, Li Y, Zhou Y, Hu X, Song C, et al. Neuroprotective effects of chlorogenic acid against apoptosis of PC12 cells induced by methylmercury. Environmental Toxicology and Pharmacology. 2008;26:13-21
  34. 34. Kwon S-H, Lee H-K, Kim J-A, Hong S-I, Kim H-C, Jo T-H, et al. Neuroprotective effects of chlorogenic acid on scopolamine-induced amnesia via anti-acetylcholinesterase and anti-oxidative activities in mice. European Journal of Pharmacology. 2010;649:210-217
  35. 35. Shafika N, Sairazi M, Sirajudeen KNS, Asari MA, Mummedy S, Muzaimi M, et al. Effect of tualang honey against KA-induced oxidative stress and neurodegeneration in the cortex of rats. BMC Complementary and Alternative Medicine. 2017;17(1):31. DOI: 10.1186/s12906-016-1534-x
  36. 36. Goes ATR, Jesse CR, Antunes MS, Lobo Ladd FV, Lobo Ladd AAB, Luchese C, et al. Protective role of chrysin on 6-hydroxydopamine-induced neurodegeneration a mouse model of Parkinson’s disease: Involvement of neuroinflammation and neurotrophins. Chemico-Biological Interactions. 2018;279:111-120. DOI: 10.1016/j.cbi.2017.10.019
  37. 37. Saad MA, Salam RMA, Kenawy SA, Attia AS. Pinocembrin attenuates hippocampal inflammation , oxidative perturbations and apoptosis in a rat model of global cerebral ischemia reperfusion. Pharmacological Reports. 2015;67:115-122. DOI: 10.1016/j.pharep.2014.08.014
  38. 38. Palla G. Isolation and identification of phenolic glucosides in liquid sugars from cane molasses. Journal of Agricultural and Food Chemistry. 1982;30:764-766. DOI: 10.1021/jf00112a035
  39. 39. Colombo R, Yariwake JH, Queiroz EF, Ndjoko K, Hostettmann K. On-line identification of minor flavones from sugarcane juice by LC/UV/MS and post-column derivatization. Journal of the Brazilian Chemical Society. 2009;20:1574-1579. DOI: 10.1590/S0103-50532009000900003
  40. 40. Jiang W, Xu Y, Zhang S, Zhu H, Hou J. Tricin 7-glucoside protects against experimental cerebral ischemia by reduction of NF- j B and HMGB1 expression. European Journal of Pharmaceutical Sciences. 2012;45:50-57. DOI: 10.1016/j.ejps.2011.10.019
  41. 41. Na CS, Hong SS, Choi YH, Lee YH, Hong SH, Lim JY, et al. Neuroprotective effects of constituents of Eragrostis ferruginea against Aβ-induced toxicity in PC12 cells. Archives of Pharmacal Research. 2010;33:999-1003. DOI: 10.1007/s12272-010-0704-5
  42. 42. Sukumaran NP, Amalraj A, Gopi S. Neuropharmacological and cognitive effects of Bacopa monnieri (L.) Wettst – A review on its mechanistic aspects. Complementary Therapies in Medicine. 2019;44:68-82. DOI: 10.1016/j.ctim.2019.03.016
  43. 43. Menghini L. A natural formulation increases brain resistance to oxidative stress. European Journal of Medicinal Plants. 2014;4:171-182. DOI: 10.9734/ejmp/2014/7150
  44. 44. Bhattacharya SK, Bhattacharya A, Kumar A, Ghosal S. Antioxidant activity of Bacopa monniera in rat frontal cortex, striatum and hippocampus. Physical Therapy Research. 2000;14:174-179. DOI: 10.1002/(SICI)1099-1573(200005)14:3<174::AID-PTR624>3.0.CO;2-O
  45. 45. Khan MB, Ahmad M, Ahmad S, Ishrat T, Vaibhav K, Khuwaja G, et al. Bacopa monniera ameliorates cognitive impairment and neurodegeneration induced by intracerebroventricular-streptozotocin in rat: Behavioral, biochemical, immunohistochemical and histopathological evidences. Metabolic Brain Disease. 2014;30:115-127. DOI: 10.1007/s11011-014-9593-5
  46. 46. Shinomol GK, Mythri RB, Srinivas Bharath MM. Bacopa monnieri extract offsets rotenone-induced cytotoxicity in dopaminergic cells and oxidative impairments in mice brain. Cellular and Molecular Neurobiology. 2012;32:455-465. DOI: 10.1007/s10571-011-9776-0
  47. 47. Uabundit N, Wattanathorn J, Mucimapura S, Ingkaninan K. Cognitive enhancement and neuroprotective effects of Bacopa monnieri in Alzheimer’s disease model. Journal of Ethnopharmacology. 2010;127:26-31. DOI: 10.1016/j.jep.2009.09.056
  48. 48. Anbarasi K, Vani G, Balakrishna K, Devi CSS. Effect of bacoside A on brain antioxidant status in cigarette smoke exposed rats. Life Sciences. 2006;78:1378-1384. DOI: 10.1016/j.lfs.2005.07.030
  49. 49. Stough C, Scholey A, Cropley V, Wesnes K, Zangara A, Pase M, et al. Examining the cognitive effects of a special extract of Bacopa monniera (CDRI 08: KeenMind): A review of ten years of research at Swinburne University. Journal of Pharmacy & Pharmaceutical Sciences. 2013;16:254-258. DOI: 10.18433/j35g6m
  50. 50. Shinomol GK, Srinivas Bharath MM. Neuromodulatory propensity of Bacopa monnieri leaf extract against 3-nitropropionic acid-induced oxidative stress: In vitro and in vivo evidences. Neurotoxicity Research. 2012;22:102-114. DOI: 10.1007/s12640-011-9303-6
  51. 51. Saraf MK, Prabhakar S, Anand A. Neuroprotective effect of Bacopa monniera on ischemia induced brain injury. Pharmacology, Biochemistry, and Behavior. 2010;97:192-197. DOI: 10.1016/j.pbb.2010.07.017
  52. 52. Limpeanchob N, Jaipan S, Rattanakaruna S, Phrompittayarat W, Ingkaninan K. Neuroprotective effect of Bacopa monnieri on beta-amyloid-induced cell death in primary cortical culture. Journal of Ethnopharmacology. 2008;120:112-117. DOI: 10.1016/j.jep.2008.07.039
  53. 53. Dhanasekaran M, Tharakan B, Holcomb LA, Hitt AR, Young KA, Manyam BV. Neuroprotective mechanisms of ayurvedic antidementia botanical Bacopa monniera. Physical Therapy Research. 2007;21:965-969. DOI: 10.1002/ptr.2195
  54. 54. Swathi G, Ramaiah CV, Rajendra W. Protective role of Bacopa monnieri against Rotenone- induced Parkinson’s disease in PC 12 cell lines. International Journal of Phytomedicine. 2017;9:219-222. DOI: 10.5138/09750185.2008
  55. 55. Ahmad MP, Hussain A, Siddiqui HH, Wahab S, Adak M. Effect of methanolic extract of Asparagus racemosus Willd. On lipopolysaccharide induced-oxidative stress in rats. Pakistan Journal of Pharmaceutical Sciences. 2015;28:509-513
  56. 56. Saxena G, Singh M, Meena P, Barber S, Sharma D, Shukla S, et al. Neuroprotective effects of Asparagus racemosus Linn root extract: An experimental and clinical evidence. Annals of Neurosciences. 2007;14:57-63. DOI: 10.5214/ans.0972.7531.2007.140302
  57. 57. Parihar MS, Hemnani T. Experimental excitotoxicity provokes oxidative damage in mice brain and attenuation by extract of Asparagus racemosus. Journal of Neural Transmission. 2004;111:1-12. DOI: 10.1007/s00702-003-0069-8
  58. 58. Bhattacharya A, Muruganandam AV, Kumar V, Bhattacharya SK. Effect of poly herbal formulation, EuMil, on neurochemical perturbations induced by chronic stress. Indian Journal of Experimental Biology. 2002;40:1161-1163
  59. 59. Bhatnagar M, Meena P, Barbar S, Joshi C. Neuroprotective response of the hippocampus region of the brain to Withania somnifera and Asparagus racemosus root extract: An in vitro study. Journal of Medicinal Plant Research. 2013;7:2259-2264. DOI: 10.5897/JMPR12.497
  60. 60. Lalert L, Kruevaisayawan H, Amatyakul P, Ingkaninan K. Neuroprotective effect of Asparagus racemosus root extract via the enhancement of brain-derived neurotrophic factor and estrogen receptor in ovariectomized rats. Journal of Ethnopharmacology. 2018;225:336-341. DOI: 10.1016/j.jep.2018.07.014
  61. 61. Pahwa P, Goel RK. Ameliorative effect of Asparagus racemosus root extract against pentylenetetrazol-induced kindling and associated depression and memory deficit. Epilepsy & Behavior. 2016;57:196, 201. DOI: 10.1016/j.yebeh.2016.01.025
  62. 62. Kulkarni SK, Verma A. Protective effect of Mentat (BR-16A), a herbal preparation on alcohol abstinence-induced anxiety and convulsions. Indian Journal of Experimental Biology. 1993;31:435-439
  63. 63. Kashyap P, Muthusamy K, Niranjan M, Trikha S, Kumar S. Sarsasapogenin: A steroidal saponin from Asparagus racemosus as multi target directed ligand in Alzheimer’s disease. Steroids. 2020;153:108529. DOI: 10.1016/j.steroids.2019.108529
  64. 64. Nandagopal M, Muralidharan P, Thirumurugan G. Cerebroprotective effect of root extract of Asparagus racemosus Willd. in global cerebral ischemia in rats. Journal of Pharmacology and Toxicology. 2011;6:49-61. DOI: 10.3923/jpt.2011.49.61
  65. 65. Joshi H, Parle M. Cholinergic basis of memory-strengthening effect of Foeniculum vulgare Linn. Journal of Medicinal Food. 2006;9:413-417. DOI: 10.1089/jmf.2006.9.413
  66. 66. Cioanca O, Hancianu M, Mircea C, Trifan A, Hritcu L. Essential oils from Apiaceae as valuable resources in neurological disorders: Foeniculum vulgare aetheroleum. Industrial Crops and Products. 2016;88:51-57. DOI: 10.1016/j.indcrop.2016.02.064
  67. 67. Nemati M, Hemmati AA, Najafzadeh H, Mansouri MT, Khodayar MJ. Evaluation the effects of Foeniculum vulgare essence on behavioral-motor disorders of Parkinson’s disease induced by reserpine in ovariectomized and non ovariectomized rats. Jundishapur Journal of Natural Pharmaceutical Products. 2018;13(1):e67391. DOI: 10.5812/jjnpp.67391
  68. 68. El Shebini SM, Abdel-Moaty M, Kazem YI, Ahmed NH, Fouad S, Mohamed MS, et al. Relation between obesity, cognition and serum amyloid β protein level and potential role of Foeniculum vulgare in reducing weight and improving cognitive functions. Journal of Biological Sciences. 2017;17:202-212. DOI: 10.3923/jbs.2017.202.212
  69. 69. Bhatti S, Ali Shah SA, Ahmed T, Zahid S. Neuroprotective effects of Foeniculum vulgare seeds extract on lead-induced neurotoxicity in mice brain. Drug and Chemical Toxicology. 2018;41:399-407. DOI: 10.1080/01480545.2018.1459669
  70. 70. Xiang X, Wu L, Mao L, Liu Y. Anti-oxidative and anti-apoptotic neuroprotectiveeffects of Azadirachta indica in Parkinson-induced functional damage. Molecular Medicine Reports. 2018;17(6):7959-7965. DOI: 10.3892/mmr.2018.8815
  71. 71. Yanpallewar S, Rai S, Kumar M, Chauhan S, Acharya SB. Neuroprotective effect of Azadirachta indica on cerebral post-ischemic reperfusion and hypoperfusion in rats. Life Sciences. 2005;76(12):1325-1338
  72. 72. Kandhare AD, Mukherjee AA, Bodhankar SL. Neuroprotective effect of Azadirachta indica standardized extract in partial sciatic nerve injury in rats: Evidencefrom anti-inflammatory, antioxidant and anti-apoptotic studies. EXCLI Journal. 2017;16:546-565. DOI: 10.17179/excli2017-161
  73. 73. Abdel Moneim AE. Azadirachta indica attenuates cisplatin-induced neurotoxicity in rats. Indian Journal of Pharmacology. 2014;46(3):316-321. DOI: 10.4103/0253-7613.132182
  74. 74. Bedi KL, Zutshi U, Chopra CL, Amla V. Picrorhiza kurroa, an ayurvedic herb, may potentiate photochemotherapy in vitiligo. Journal of Ethnopharmacology. 1989;27(3):347-352
  75. 75. Simonyi A, Serfozo P, Lehmidi TM, Cui J, Gu Z, Lubahn DB, et al. The neuroprotective effects of apocynin. Frontiers in Bioscience (Elite Edition). 2012;4:2183-2193
  76. 76. Marín M, Giner RM, Ríos JL, Recio MC. Protective effect of apocynin in a mouse model of chemically-induced colitis. Planta Medica. 2013;79(15):1392-1400. DOI: 10.1055/s-0033-1350710
  77. 77. Romanini CV, Ferreira ED, Soares LM, Santiago AN, Milani H, de Oliveira RM. 4-Hydroxy-3-methoxy-acetophenone-mediated long-lasting memory recovery, hippocampal neuroprotection, and reduction of glial cell activation after transient global cerebral ischemia in rats. Journal of Neuroscience Research. 2015;93(8):1240-1249. DOI: 10.1002/jnr.23575
  78. 78. Zahiruddin S, Khan W, Nehra R, Alam MJ, Mallick MN, Parveen R, et al. Pharmacokinetics and comparative metabolic profiling of iridoid enriched fraction of Picrorhiza kurroa - An Ayurvedic herb. Journal of Ethnopharmacology. 2017;197:157-164. DOI: 10.1016/j.jep.2016.07.072
  79. 79. Kim N, Do J, Ju IG, Jeon SH, Lee JK, Oh MS. Picrorhiza kurroa prevents memory deficits by inhibiting NLRP3 Inflammasome activation and BACE1 expression in 5xFAD mice. Neurotherapeutics. 2020;17(1):189-199. DOI: 10.1007/s13311-019-00792-7
  80. 80. Morikawa T, Nakanishi Y, Inoue N, Manse Y, Matsuura H, Hamasaki S, et al. Acylated iridoid glycosides with hyaluronidase inhibitory activity from the rhizomes of Picrorhiza kurroa Royle ex. Benth. Phytochemistry. 2020;169:112185. DOI: 10.1016/j.phytochem.2019.112185
  81. 81. Dinda B, Dinda M, Kulsi G, Chakraborty A, Dinda S. Therapeutic potentials of plant iridoids in alzeimer’s and parkinson’s diseases. A review. European Journal of Medicinal Chemistry. 2019;169:185-199
  82. 82. Tavakkoli A, Iranshahi M, Hasheminezhad SH, Hayes AW, Karimi G. The neuroprotective activities of natural products through the Nrf2 upregulation. Phytotherapy Research. 2019;33(9):2256-2273. DOI: 10.1002/ptr.6427
  83. 83. Yuan NN, Cai CZ, Wu MY, Su HX, Li M, Lu JH. Neuroprotective effects of berberine in animal models of Alzheimer’s disease: A systematic review of pre-clinical studies. BMC Complementary and Alternative Medicine. 2019;19(1):109. DOI: 10.1186/s12906-019-2510-z
  84. 84. Soudi SA, Nounou MI, Sheweita SA, Ghareeb DA, Younis LK, El-Khordagui LK. Protective effect of surface-modified berberine nanoparticles against LPS-induced neurodegenerative changes: A preclinical study. Drug Delivery and Translational Research. 2019;9(5):906-919. DOI: 10.1007/s13346-019-00626-1
  85. 85. Maleki SN, Aboutaleb N, Souri F. Berberine confers neuroprotection in coping with focal cerebral ischemia by targeting inflammatory cytokines. Journal of Chemical Neuroanatomy. 2018;87:54-59. DOI: 10.1016/j.jchemneu.2017.04.008
  86. 86. Haghani M, Shabani M, Tondar M. The therapeutic potential of berberine against the altered intrinsic properties of the CA1 neurons induced by Aβ neurotoxicity. European Journal of Pharmacology. 2015;758:82-88. DOI: 10.1016/j.ejphar.2015.03.016
  87. 87. Lin X, Zhang N. Berberine: Pathways to protect neurons. Phytotherapy Research. 2018;32(8):1501-1510. DOI: 10.1002/ptr.6107
  88. 88. Fan D, Liu L, Wu Z, Cao M. Combating neurodegenerative diseases with the plant alkaloid Berberine: Molecular mechanisms and therapeutic potential. Current Neuropharmacology. 2019;17(6):563-579. DOI: 10.2174/1570159X16666180419141613
  89. 89. Hao F, Jia L-H, Li X-W, Zhang Y-R, Liu X-W. Garcinol upregulates GABAA and GAD65 expression, modulates BDNF-TrkB pathway to reduce seizures in pentylenetetrazole (PTZ)-induced epilepsy. Medical Science Monitor. 2016;22:4415-4425
  90. 90. Weng M-S, Liao C-H, Yu S-Y, Lin J-K. Garcinol promotes neurogenesis in rat cortical progenitor cells through the duration of extracellular signal-regulated kinase signaling. Journal of Agricultural and Food Chemistry. 2011;59:1031-1040. DOI: 10.1021/jf104263s
  91. 91. Balasubramanyam K, Altaf M, Varier RA, Swaminathan V, Ravindran A, Sadhale PP, et al. Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression. The Journal of Biological Chemistry. 2004;279:33716-33726. DOI: 10.1074/jbc
  92. 92. Hegarty SV, Sullivan AM, O’Keeffe GW. The Epigenome as a therapeutic target for Parkinson’s disease. Neural Regeneration Research. 2016;11:1735-1738. DOI: 10. 4103/1673-5374.194803
  93. 93. Mazumder MK, Paul R, Phukan BC, Dutta A, Chakrabarty J, Bhattacharya P, et al. Garcinol, an effective monoamine oxidase-B inhibitor for the treatment of Parkinson’s disease. Medical Hypotheses. 2018;117:54-58. DOI: 10.1016/j.mehy.2018.06.009
  94. 94. Ryu Y-K, Park H-Y, Go J, Kim Y-H, Hwang JH, Choi D-H, et al. Effects of histone acetyltransferase inhibitors on l-DOPA-induced dyskinesia in a murine model of Parkinson’s disease. Journal of Neural Transmission. 2018;125:1319-1331. DOI: 10.1007/s00702-018-1902-4
  95. 95. Bastide MF, Dovero S, Charron G, Porras G, Gross CE, Fernagut P-O, et al. Immediate-early gene expression in structures outside the basal ganglia is associated to L-DOPA-induced dyskinesia. Neurobiology of Disease. 2014;62:179-192. DOI: 10.1016/j.nbd.2013.09.020
  96. 96. Antala BV, Patel MS, Bhuva SV, Gupta S, Rabadiya S, Lahkar M. Protective effect of methanolic extract of Garcinia indica fruits in 6-OHDA rat model of Parkinson’s disease. Indian Journal of Pharmacology. 2012;44:683-687. DOI: 10.4103/0253-7613.103242
  97. 97. Park G, Tan J, Garcia G, Kang Y, Salvesen G, Zhang Z. Regulation of histone acetylation by autophagy in Parkinson disease. The Journal of Biological Chemistry. 2016;291:3531-3540. DOI: 10.1074/jbc.M115.675488
  98. 98. Deb S, Banashree C, Phukan M, Mazumder K, Duttaa A, Paul R, et al. Garcinol, a multifaceted sword for the treatment of Parkinson’s disease. Neurochemistry International. 2019;128:50-57
  99. 99. Dhingra D, Goyal PK. Inhibition of MAO and GABA: Probable mechanisms for antidepressant-like activity of Nardostachys jatamansi DC. in mice. Indian Journal of Experimental Biology. 2008;46(4):212-2018
  100. 100. Salim S, Ahmad M, Zafar KS, Ahmad AS, Islam F. Protective effect of Nardostachys jatamansi in rat cerebral ischemia. Pharmacology, Biochemistry, and Behavior. 2003;74(2):481-486
  101. 101. Ahmad M, Yousuf S, Khan MB, Hoda MN, Ahmad AS, Ansari MA, et al. Attenuation by Nardostachys jatamansi of 6-hydroxydopamine-induced parkinsonism in rats: Behavioral, neurochemical, and immunohistochemical studies. Pharmacology, Biochemistry, and Behavior. 2006;83(1):150-160
  102. 102. Joshi H, Parle M. Nardostachys jatamansi improves learning and memory in mice. Journal of Medicinal Food. 2006;9(1):113-118
  103. 103. Lyle N, Bhattacharyya D, Sur TK, Munshi S, Paul S, Chatterjee S, et al. Stress modulating antioxidant effect of Nardostachys jatamansi. Indian Journal of Biochemistry & Biophysics. 2009;46(1):93-98
  104. 104. Shah J, Goyal R. Investigation of neuropsychopharmacological effects of a polyherbal formulation on the learning and memory process in rats. Journal of Young Pharmacists. 2011;3(2):119-124. DOI: 10.4103/0975-1483.80296
  105. 105. Khan MB, Hoda MN, Ishrat T, Ahmad S, Moshahid Khan M, Ahmad A, et al. Neuroprotective efficacy of Nardostachys jatamansi and crocetin in conjunction with selenium in cognitive impairment. Neurological Sciences. 2012;33(5):1011-1020. DOI: 10.1007/s10072-011-0880-1
  106. 106. Suryavanshi S, Raina P, Deshpande R, Kaul-Ghanekar R. Nardostachys jatamansi root extract modulates the growth of IMR-32 and SK-N-MC neuroblastoma cell lines through MYCN mediated regulation of MDM2 and p53. Pharmacognosy Magazine. 2017;13(49):21-24. DOI: 10.4103/0973-1296.197645
  107. 107. Yoon CS, Kim DC, Park JS, Kim KW, Kim YC, Oh H. Isolation of novel Sesquiterpeniods and anti-neuroinflammatory metabolites from Nardostachys jatamansi. Molecules. 2018;23(9):E2367. DOI: 10.3390/molecules23092367
  108. 108. Kim KW, Yoon CS, Kim YC, Oh H. Desoxo-narchinol a and Narchinol B isolated from Nardostachys jatamansi exert anti-neuroinflammatory effects by up-regulating of nuclear transcription factor Erythroid-2-related factor 2/Heme Oxygenase-1 Signaling. Neurotoxicity Research. 2019;35(1):230-243. DOI: 10.1007/s12640-018-9951-x
  109. 109. Singh GK, Rai G, Chatterjee SS, Kumar V. Effects of ethanolic extract of Fumaria indica L. on rat cognitive dysfunctions. AYU. 2013;34(4):421-429. DOI: 10.4103/0974-8520.127727
  110. 110. Singh GK, Chauhan SK, Rai G, Chatterjee SS, Kumar V. Potential antianxiety activity of Fumaria indica: A preclinical study. Pharmacognosy Magazine. 2013;9(33):14-22. DOI: 10.4103/0973-1296.108129
  111. 111. Asghari MH, Fallah M, Moloudizargari M, Mehdikhani F, Sepehrnia P, Moradi B. A systematic and mechanistic review on the Phytopharmacological properties of Alhagi species. Ancient Science of Life. 2016;36(2):65-71. DOI: 10.4103/asl.ASL_37_16
  112. 112. Muhammad G, Hussain MA, Anwar F, Ashraf M, Gilani AH. Alhagi: A plant genus rich in bioactives for pharmaceuticals. Phytotherapy Research. 2015;29(1):1-13. DOI: 10.1002/ptr.5222
  113. 113. Singh VP, Yadav B, Pandey VB. Flavanone glycosides from Alhagi pseudalhagi. Phytochemistry. 1999;51(4):587-590
  114. 114. Ghosal S, Srivastava RS, Bhattacharya SK, Debnath PK. The active principles of Alhagi pseudalhagi: Beta-phenethylamine and tetrahydroisoquinoline bases. Planta Medica. 1974;26(4):318-326
  115. 115. Mundugaru R, Sivanesan S, Popa-Wagner A, Udaykumar P, Kirubagaran R, Kp G, et al. Pluchea lanceolata protects hippocampal neurons from endothelin-1 induced ischemic injury to ameliorate cognitive deficits. Journal of Chemical Neuroanatomy. 2018;94:75-85. DOI: 10.1016/j.jchemneu.2018.09.002
  116. 116. Mundugaru R, Sivanesan S, Udaykumar P, Rao N, Chandra N. Protective effect of Pluchea lanceolata against aluminum chloride-induced neurotoxicity in Swiss albino mice. Pharmacognosy Magazine. 2017;13(Suppl 3):S567-S572. DOI: 10.4103/pm.pm_124_17
  117. 117. Srivastava P, Mohanti S, Bawankule DU, Khan F, Shanker K. Effect of Pluchea lanceolata bioactives in LPS-induced neuroinflammation in C6 rat glial cells. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2014;387(2):119-127. DOI: 10.1007/s00210-013-0924-6
  118. 118. Patel NG, Patel KG, Patel KV, Gandhi TR. Validated HPTLC method for quantification of luteolin and apigenin in Premna mucronata Roxb., Verbenaceae. Advances in Pharmacological Sciences. 2015;2015:682365. DOI: 10.1155/2015/682365
  119. 119. Shukla SD, Jain S, Sharma K, Bhatnagar M. Stress induced neuron degeneration and protective effects of Semecarpus anacardium Linn. And Withania somnifera Dunn. in hippocampus of albino rats: An ultrastructural study. Indian Journal of Experimental Biology. 2000;38(10):1007-1013
  120. 120. Devi SJR, Kumar BP. In silico screening for anti-inflammatory bioactive molecules from ayurvedic decoction, Balagulchyadi Kashayam. Current Computer-Aided Drug Design. 2019. DOI: 10.2174/1573409915666191015113753
  121. 121. Birla H, Rai SN, Singh SS, Zahra W, Rawat A, Tiwari N, et al. Tinospora cordifolia suppresses neuroinflammation in parkinsonian mouse model. Neuromolecular Medicine. 2019;21(1):42-53. DOI: 10.1007/s12017-018-08521-7
  122. 122. Sharma A, Kaur G. Tinospora cordifolia as a potential neuroregenerative candidate against glutamate induced excitotoxicity: An in vitro perspective. BMC Complementary and Alternative Medicine. 2018;18(1):268. DOI: 10.1186/s12906-018-2330-2336
  123. 123. Singh H, Kaur T, Manchanda S, Kaur G. Intermittent fasting combined withsupplementation with Ayurvedic herbs reduces anxiety in middle aged female rats by anti-inflammatory pathways. Biogerontology. 2017;18(4):601-614. DOI: 10.1007/s10522-017-9706-8
  124. 124. Bhattacharya S, Haldar PK. Neuropharmacological properties of Trichosanthes dioica root. Chinese Journal of Natural Medicines. 2013;11(2):158-163. DOI: 10.1016/S1875-5364(13)60043-6

Written By

Uma Ranjan Lal and Snigdha Lal

Submitted: 18 October 2019 Reviewed: 08 March 2020 Published: 07 May 2020