Abstract
Traditional Indian medicine (Ayurveda) has a long history of treating central nervous system (CNS) disorders. Ayurvedic plants are typically rich in flavonoids, lignans, sterols, tannins and alkaloids that exert potent anti-inflammatory, anti-oxidant, neuroprotective and neurotropic effects in both humans and rodents. The zebrafish (Danio rerio) has emerged as a useful model species and a powerful vertebrate in vivo system for CNS disease modeling and drug screening. As zebrafish continue to demonstrate their growing utility for studying a wide range of pharmaceutical therapies, including Chinese and American traditional medicines, here we discuss CNS effects of various Ayurvedic medicinal plants, and how they can be further elucidated using zebrafish models. We also evaluate the existing challenges and limitations of using zebrafish models for studying Ayurvedic pharmacotherapy, as well as outline future directions of translational research in this field.
Keywords: Ayurveda, Brain disorders, Zebrafish, Treatment, Novel drugs
1. Introduction
Traditional Indian medicine, Ayurveda, has been in use for more than 5000 years [1], commonly applied in the region to treat various human diseases, including cancer, infectious, metabolic, cardiovascular and neurological disorders [2]. Ayurveda (meaning “The Science of Life” in Sanskrit) uses native plants rich in flavonoids, lignans, sterols, tannins and alkaloids, with robust anti-inflammatory, anti-arthritic, antioxidant and other beneficial properties [3,4]. Today, Ayurveda remains one of the oldest and most commonly used therapies globally, whose popularity is also expanding. For example, the Ayurvedic medicines have a long history of use and relatively few side effects, hence contributing to its growing consumption, with nearly 90 % Indians and up to 10 % American adults presently treated by Ayurvedic medicines.
Numerous Ayurvedic plants (e.g., curcumin, Indian pennywort, Indian ginseng, tulsi) have long been used to treat various brain disorders [4], known as ‘vata-vyadhi’ in Sanskrit (from ‘vata’- energy moving from brain to nerves to control human body, and ‘vyadhi’, the disease) [2]. Traditionally, Ayurveda recognizes three main energies (doshas) of the body - pitta (metabolism), kapha (structure) and vata (mental). Set at birth at various ratios to maintain physiological homeostasis, their imbalance is thought to cause mental illnesses [5,6]. Among the three doshas, one is typically dominant in each patient, and such individual constitution (prakriti) determines the susceptibility to specific disorders, based on physical, psychological, physiological and behavioral traits that are independent of social, ethnic and geographical variables [7].
For millennia, Ayurveda has successfully treated various human central nervous system (CNS) disorders, including anxiety, depression, cognitive decline, as well as Alzheimer's (AD), Huntington's (HD) and Parkinson's diseases (PD, Tables 1 and 1-Supplementary material). Complementing clinical studies, experimental animal models, especially laboratory rodents, are increasingly utilized in translational Ayurveda research [6], including studying CNS effects of Ayurvedic medicine (see further). Mounting evidence also suggests that alternative model species, such as zebrafish (Danio rerio Hamilton), can also be a useful tool to study effects of Ayurvedic medicines on CNS functions [8,9]. To examine this further, here we discuss how CNS effects of various Ayurvedic medicinal plants can be elucidated using zebrafish models, based on a comprehensive search of published literature from all major biomedical databases (Web of Science, PubMed, ScienceDirect, Scopus, and Google Scholar). We also evaluate the existing challenges and limitations of using fish models for studying Ayurvedic pharmacotherapy, as well as outline future directions of translational research in this field.
Table 1.
Selected Ayurvedic herbs, their active compounds and various CNS effects.
Ayurvedic plants | Botanical name | Major active compounds | Targeted brain conditions | References |
---|---|---|---|---|
Curcumin | Curcuma longa | Curcumin | Alzheimer's (AD) and Parkinson's (PD) diseases, anxiety | [2,3] |
Brahmi | Bacopa monnieri | Bacoside A, bacoside B and bacopasaponins, D-mannitol | AD, PD, anxiety and stress | [21,22] |
Ashwagandha | Withania somnifera | Withanolides A, B, Q and IV, V, somnine, somniferine and sominone | AD, PD and Huntington's disease (HD), stress and epilepsy | [11,14] |
Shatavari | Asparagus racemosus | Shatavarin IV | AD, PD and neuroinflammation | [2] |
Shankhpushpi | Convolvulus prostratus | Glycosides, flavonol, anthocyanins and steroids | Anxiety, mental fatigue, and insomnia | [97,98] |
Jatamansi | Nardostachys jatamansi | Sesquiterpenoids and valeriananoids | AD and insomnia | [99,100] |
Mandukaparni | Centella asiatica | Asiaticoside A, B and asiatic acid | AD and PD | [101] |
Gugul | Commiphora wightii | Guggulsterol (I-III, IV, V) and E,Z-guggulsteron | Dementia | [102] |
Please see text and Table 1-Supplementary material for a complete list of Ayurvedic plants mentioned here.
2. Selected examples of experimental models to study CNS effects of Ayurvedic medicine
As already noted, in addition to rich clinical data, multiple animal (experimental) models have been used to study Ayurvedic medicines [6]. For example, Indian ginseng (Withania somnifera Linnaeus) that is efficient in managing stress, epilepsy and PD clinically [10], exhibits positive effect in experimental AD models [2], reducing neuro-fibrillary tangles and scavenging free radicals to protect dopaminergic neurons in rodents [11]. Known as ashwagandha and used in Ayurveda for millennia to treat pain, inflammation, insomnia and stress, this plant provides an extract that reduces lipid peroxidation, upregulates tyrosine hydroxylase (TH) in substantia nigra and exerts anti-oxidant properties in mice [10,12]. In transgenic AD mouse model, its root extracts show neuroprotective effects against amyloid beta (Aβ) and peroxide toxicity, and decrease Aβ formation in the brain [13]. Promoting neurite regeneration and increasing central gamma-aminobutyric acid (GABA) tone, this plant is beneficial in epilepsy [14], also improving cognition, restoring motor function and controlling acetylcholine esterase (AChE) activity in an experimental HD model [15]. Methanolic and aqueous extracts of Indian ginseng also promote serotonergic transmission via the 5HT2A/2C receptor in the mouse model of the obsessive compulsive disorder [16].
Another popular plant, water hyssop (Bacopa monnieri Linnaeus Wettstein), known in Ayurveda as brahmi (‘plant of Lord Brahma’), is widely used to treat cognitive deficits, anxiety and epilepsy clinically [17]. This plant contains several saponins (bacoside A3, bacopaside II, bacopasaponin C and bacopaside I) that determine its therapeutic effects and biological activity in vivo. For example, recommended for human AD and PD patients [18], water hyssop also improves cognition and reduces anxiety and stress in animal models [5], and promotes neuroplasticity by increasing the expression of brain-derived neurotropic factor (BDNF) in mice [19]. Lower dose of this plant extract reduces inflammatory interleukins in microglial cells [20], also decreasing oxidative stress, protecting neuronal and glial cells, and lowering neuro-inflammation in rats [21,22], in addition to its well-reported cognitive enhances activity in humans [17].
Curcumin is an active compound of turmeric (Curcuma longa Linnaeus), promoting neuroprotection in AD and exerting anti-inflammatory, anti-oxidant (and anxiolytic) activity in mouse models and in vitro [3]. In Ayurveda, this plants is used as an anti-amyloidogenic and anti-inflammatory agent, also showing neuroprotective action in tardive dyskinesia, AD, depression, epilepsy and other CNS disorders [23]. Inhibiting monoamine oxidase (MAO)-A in mice, curcumin elevates brain monoamines [24] in synergistic interactions with serotonergic antidepressants [25]. Currently approved by the US Food and Drug Administration (FDA) in Phase II clinical trials against AD as Longvida [26], it also reduces the activation of microglia and astrocytes, as well as cytokine production and the nuclear factor kappa-light-chain enhancer of activated B cells (NFκB) signaling pathway, all suggesting beneficial activity against AD, in addition to its clinical use as a complementary treatment for neuropathic pain [27].
Indian pennywort (Centella asiatica Linnaeus) is another commonly used Ayurvedic plant [28] with well-known potent antimicrobial, antioxidant, antiulcer, anti-diabetic, anticancer, antidepressant and nootropic clinical activity [29]. Mentioned in Ayurveda as gotu kola or mandukaparni, and rich is flavonoids, antioxidants and vitamins, Indian pennywort has been traditionally used to improve mindfulness and cognitions, as well as an antiepileptic, antidepressant, wound healing and antiseptic therapy. Representing a major Ayurvedic ‘Medhya Rasayana’ (literally - ‘improving mind’, nootropic) plant, it also improves memory and learning in rodents [30,31], inhibits phospholipase A2 (PL-A2) and acetylcholine esterase (AChE), protects cerebellum from Aβ-induced neurotoxicity [32], evokes anti-inflammatory activity in lipopolysaccharide (LPS)-treated rat neurons, and possesses anxiolytic-, anti-stress and antidepressant-like effects in vivo [33].
Morning glory (Convolvulus pluricaulis Linnaeus, another major Medhya Rasayana/nootropic Ayurvedic plant known as shankhapushpi) and spikenard (Nardostachys jatamansi Linnaeus, a promnestic, antidepressant and antiepileptic plant) both exert nootropic effects clinically [2] and in rodent models [34]. Likewise, Asian pigeonwing (Clitoria ternatea Linnaeus) root extract improves rat passive avoidance learning and retention. A traditional Ayurvedic medicine aparajita, it has long been used as a nootropic, anxiolytic, antidepressant, anticonvulsant and sedative agent. Commonly known as ‘butterfly pea’, this plant generates multiple metabolites (e.g., triterpenoids, glycosides, anthocyanins and steroids) that contribute to its biological activity in vivo [35].
Considered one of the most important Ayurvedic herbs, tulsi (Ocimum sanctum Linnaeus) promote general rejuvenation of mind and body, improves cognition and metabolism [36], reduces stress, anxiety and depression [37], and exerts anticonvulsant activity clinically [38]. Similarly to humans, ethanol extract of tulsi evokes an anti-stress profile in rodents, reducing plasma corticosterone responses to acute or chronic noise stress, and improving memory [39]. The lipid peroxidase activity, deficits in active avoidance learning and retention of learned behavior are reversed by tulsi extract in the rat AD model, also normalizing superoxide dismutase activity [40].
The role of brain–gut axis has been increasingly recognized in pathogenesis of various CNS disorders, including depression, anxiety and neurodegenerative disorders [41]. For example, the irritable bowel disease (IBD), causing a chronic inflammation of the gastrointestinal tract and causing both Crohn disease and ulcerative colitis), also increases CNS inflammatory responses, further contributing to affective disorders clinically [42]. Crohn patients have higher anxiety scores than healthy individuals [43], and are more prone to depression [44]. Various Ayurvedic medicines have long been used to correct the brain-gut axis clinically [45]. For instance, the curcumin extract (Curcugen) administered for 8 weeks at a dose of 500 mg once daily is associated with greater improvements in digestive complaints and anxiety levels in adults with self-reported digestive complaints [46]. Subjects treated with another Ayurvedic medicine, ashwagandha (Withania somnifera L. Dunal), for 8 weeks demonstrate significant improvements compared with the placebo group in cognitive evaluation (e.g., immediate and general memory) [47]. In addition to clinical finding, rodent models show reduced anxiety- and depression-like behavioral deficits following their treatment with Ayurvedic medicine (e.g., Withania somnifera [48], aqueous extract of Azadirachta indica A. Juss [49], probiotics associated with curcumin [50]). Collectively, these findings strongly support the growing value of animal models for studying various aspects of CNS effects of Ayurvedic medicine.
3. Assessing selected Ayurvedic medicines in zebrafish CNS models
Complementing clinical and rodent findings, the zebrafish has recently emerged as a powerful vertebrate in-vivo system for CNS disease modeling and drug screening [51]. For example, both adult and larval zebrafish represent powerful vertebrate in-vivo model systems to study CNS functions [52] due to their high genetic and physiological similarity to humans, rapid development and shared neurotransmitter systems [53]. Other advantages of zebrafish models include neuromorphological similarity to humans [54], easy maintenance, high fecundity [55] and the availability of well-established behavioral assays. Zebrafish are also sensitive to all major classes of CNS drugs, offer cost-effective model for in vivo drug screening and show potential for high-throughput screening (HTS) [56].
Recognizing its potential for bioscreening various medicines, the zebrafish has recently become utilized as a preclinical model for obtaining insights into molecular mechanisms of action and for developing novel treatments for neurological disorders based on Ayurvedic herbal medicine [57]. Mounting evidence, summarized in Table 2 and including selected examples discussed further in detail, strongly supports the utility of zebrafish in studying CNS effects of Ayurvedic plants. Fig. 1 depicts examples of CNS effects.
Table 2.
Selected examples of using zebrafish models for screening CNS effects of Ayurvedic medicines.
Medicines | Botanical name | Sex | CNS effects | References |
---|---|---|---|---|
Indian pennywort | Centella asiatica | Both | Protected dopaminergic neurons from rotenone toxicity by increasing BDNF levels and reduced α-synuclein aggregation | [28,103] |
Indian ginseng | Withania somnifera | Both | Improved brain antioxidant status and neuroprotection against benzo[a] pyrene toxicity | [60] |
Suvarna Bhasmaa | - | Not specified | Reduced behavioral deficits induced by rotenone administration (a PD model) | [70] |
- | Both | Reduced anxiety-like behavior | [71] | |
Curcumin | Curcuma longa | Both | Improved locomotion and reduced rotenone toxicity in PD model | [61] |
Tulsi | Ocimum tenuiflorum | Male | Protected memory against scopolamine | [63] |
Asian pigeonwing | Clitoria ternatea | Both | Reduced stress responses induced by reserpine | [65] |
Brahmi | Bacopa monnieri | Not specified | Reduced anxiety-like behavior and cortisol levels following exposure to stress stimuli | [66] |
Carob | Ceratonia siliqua | Both | Antioxidant and anti-AChE activity, improved cognitive function in the 6-OHDA-induced PD model | [62] |
Neem | Azadirachta indica | Not specified | Reduced locomotion and increased anxiety-like behavior | [104] |
gold nanoparticles.
Fig. 1.
Selected examples of CNS effects of Ayurvedic medicinal plants in zebrafish models.Ceratonia siliqua L. (carob) is a Mediterranean medicinal plant prevents the cognitive impairment of 6-hydroxydopamine (6-OHDA, a Parkinson's disease (PD) model) [62]. Ashwagandha (Withania somnifera Dunal) leaf extract ameliorated the increase in the pyknotic neuronal counts in periventricular gray zone caused by benzo[a]pyrene neurotoxicity [60]. The extract of tulsi leaves reduces memory deficits caused by scopolamine in zebrafish [63]. Curcumin, the main ingredient of turmeric, protects against the locomotor impaired caused by rotenone (a PD-like model) [61].
3.1. Indian pennywort
Indian pennywort has been extensively tested in zebrafish. For example, in rotenone neurotoxicity assay, when co-incubated with the extract of Indian pennywort, adult zebrafish elevate BDNF levels in midbrain, their dopaminergic neurons become protected from toxicity, and fish display reduced PD-like phenotypes (e.g., lower alpha-synuclein aggregation, increased locomotion and higher brain dopamine levels) [28]. The active constituents of this herb (e.g., brahmoside and brahminoside) are traditionally used for CNS relaxation in humans and exert similar effects in animals [58], whereas asiaticoside, its another active compound, reduces hydrogen peroxide-induced cell death, free radical concentrations in-vitro and Aβ levels in rat hippocampus [59].
3.2. Indian ginseng
Indian ginseng extract exposure attenuates benzo[a]pyrene neurotoxicity in adult zebrafish, increasing brain antioxidant status and restoring normal scototaxic (dark preference) behavior [60]. Interestingly, in Ayurveda practice, this plant is commonly used as an anxiolytic, nootropic [11], anti-PD/AD/HD and anti-epileptic agent [10], and its effects in zebrafish strikingly parallel clinical findings.
3.3. Curcumin and carob
Curcumin improves locomotor performance in adult zebrafish rotenone-based PD models [61], whereas leaf extract of tannins-rich carob (St. John's bread, Ceratonia siliqua Linnaeus) improves cognitive functions and lowers anxiety in adult zebrafish PD model evoked by a neurotoxin 6-hydroxydopamine (6-OHDA) – the effects likely mediated by restoring brain antioxidant status and regulating AChE activity [62].
3.4. Tulsi and areca palm
Paralleling clinical nootropic use of tulsi, its leaf extract reduces memory impairment in adult zebrafish caused by scopolamine, an anticholinergic drug commonly used to induce memory deficits [63]. Known as puga in Ayurveda, extracts from leaves and nuts of areca palm (Areca catechu Linnaeus) are commonly used clinically to treat ulcers, pain and indigestions. Arecoline, the main psychoactive compound of areca (betel) nuts and their water extract, have been recently studied in zebrafish, whose CNS responses parallel clinical and rodent findings, and show pronounced anxiolytic-like behavior and elevated norepinephrine and serotonin levels [64]. On the one hand, by showing similar effects across taxa, these data further support evolutional conservation of psychoactive and neurochemical effects of Ayurvedic medicines across vertebrates. On the other hand, although anxiolytic effects of puga are not listed as its major use in Ayurveda, the fact that consistent zebrafish, rodent and clinical CNS phenotypes are evoked by arecoline [64] suggests that repurposing Ayurvedic medicines, for example, based on their zebrafish-based CNS screening, may also lead to novel potential pharmacotherapies for various brain disorders.
3.5. Asian pigeonwing
Asian pigeonwing, a potent stress-reliever and brain stimulant in humans and rat models, has also been tested in zebrafish [65]. In the fish model of pharmacogenic affective syndrome, the clitorienolactone- and isoflavonoid-rich extract from the roots of this plant, reduces motor deficits (freezing and hypolocomotion) induced by a dopamine-depleting drug reserpine [65], hence strikingly paralleling brain stimulant action of this popular Ayurvedic medicine in humans.
3.6. Water hyssop
Water hyssop, traditionally used in Ayurveda as a neural tonic, potently reduces thigmotaxic and scototaxic behavior (anxiolytic effect) in zebrafish larvae exposed to osmotic stress as compared to untreated stressed larvae, as well as reduced the stress-induced release of cortisol in zebrafish larvae, suggesting that this plant may be an option for managing stress and anxiety [66].
3.7. Satawari and black myrobalan
Given the success of Ayurvedic medicines in targeting the brain–gut axis [45], and reflecting the growing potential of zebrafish in modeling this axis [67], some compounds have been tested in zebrafish models, aiming to better understand the gut-brain axis, as well as its influence on CNS disorders. For example, in a zebrafish model of IBD, methanol extracts of Indian plants water roof (Asparagus racemosus Willdstein) and especially black myrobalan (Terminalia chebula Retzius), known as shatavari and haritaki in Ayurveda, lower inflammatory cells in blood and improve pathological scores in the gut compared with positive prednisolone control [68]. In sum, these findings support zebrafish as useful model species to study the effects of Ayurvedic medicinal plants on inflammatory and CNS disorders, as well as the development of novel therapies for targeting gut-brain axis [67].
3.8. Suvarna Bhasma
In addition to various herbal medicines, other types of Ayurvedic treatments can also be tested in zebrafish. For instance, believed to have rejuvenating, nootropic and stress-reducing properties in Ayurveda, nano-sized gold particles, Suvarna Bhasma (SB), have a powerful potential to treat neurological diseases (e.g., PD) clinically [69]. In a zebrafish PD model (induced by rotenone administration), behavioral deficits are reduced by SB treatment, which also prevents dopamine deficits in fish brain, paralleling neuroprotective effects of SB against PD [70]. Furthermore, utilizing non-toxic doses of SB in zebrafish produced beneficial (anxiolytic-like) behavioral effects in the novel tank test, also strikingly resembling well-established calming, anti-stress effects of this medicine in humans [71].
4. Prospects of zebrafish models in neuroscience and Ayurvedic drug screening research
Analyses of reported CNS effects of Ayurvedic plants in animal models (Table 1-Supplementary material) reveal predominantly promnestic (nootropic) and anti-stress effects in vivo, which are generally in line with clinical disorders treated with Ayurvedic herbs (Table 1). However, identifying novel clinical effects of traditional Ayurvedic plants may also be necessary, especially targeting other illnesses, such as neurodevelopmental disorders like autism spectrum disorder, currently lacking the specific drug therapy [72]. Ayurvedic drugs have already shown its efficacy in the treatment of pain disorders as strong analgesic and anti-inflammatory agents [73], and may also be used to treat complex multifaceted brain disorders, such as schizophrenia [74], where, for example, water hyssop extracts given chronically improve positive and negative clinical symptoms of psychoses without overt adverse effects [75]. As zebrafish become a valuable model organism to mimic schizophrenia [76] and other related CNS conditions, testing this possibility in fish becomes possible.
Importantly, the effectiveness of Ayurvedic medicinal plants depends on the bioavailability of their active compounds over a sustained period of time [77]. Addressing this aspect, recent nanotechnologies have been developed to enhance bioavailability and bioactivity of Ayurvedic medicines [77]. For instance, micro and nano-carrier-mediated drug delivery systems [78] can prolong biological effects of the drugs in the action site by reducing its clearance and increasing therapeutic effects [79]. Thus, capitalizing on home-tank water immersion-based drug administration, similar long-term treatment approaches can be developed in zebrafish, to further enhance their CNS models and screens for Ayurvedic medicines. The supercritical fluid micronization technology represents another useful tool to increase drug bioavailability [80] and therapeutic efficacy [81]. For example, micronized resveratrol shows promising effects in an epilepsy model, preventing tonic/clonic seizures in adult zebrafish [82]. Similar approaches can also be used to enhance therapeutic effects of Ayurvedic medicines in this and other CNS disease models using zebrafish.
Various intrinsic factors (e.g., sex, individual, anatomic, behavioral and physiological differences) modulate pharmacological responses in clinical and preclinical studies [83]. For instance, as body temperature modulates drug effects in humans and rodents [84], this factor can differentially impact Ayurvedic therapies in poikilothermic zebrafish (typically maintained in laboratories at 25–28 °C) and homeothermic mammals (typically maintaining a 35–37 °C body temperature). Some extrinsic factors can also differentially influence behavioral and CNS drug response in preclinical studies across species, as, for example, experimenter's sex impacts such responses in rodents [85], but not in zebrafish [86].
For large-scale Ayurveda studies, zebrafish high-throughput imaging assay can be developed for drug neurotoxicity screening, to better understand drug pharmacokinetics and pharmacodynamics. For instance, Ayurveda utilizes a tedious procedure for the preparation of rasasindura (a mercury-based medicinal formulation), which in zebrafish larvae, even at high concentrations of 1000 ppm, did not show toxicity or morphological changes, suggesting that this Ayurvedic medicine can be non-toxic in vivo up to a reasonable concentration [87]. While many experiments with Ayurvedic drugs involved adult fish (Table 2), zebrafish larvae is another promising tool since their small size enables placing them into multi-well plates filled with only 200 μl of fluid, requiring few milligrams of compounds for screening [88]. Although identification of bioactive compounds from Ayurvedic plants normally requires multiple chromatographic procedures and large amounts of material, which is problematic in larval assays, novel methods for larval behavioral testing with micro-fractionation techniques for bioactive compound identification from traditional herbal medicine have been developed [89].
An interesting line of research also involves the rasa shastra branch of Ayurveda that combines traditional Indian medicinal plants with inorganic components, mostly metals. For example, the combination of Ayurvedic plant extracts with metals may have some therapeutic potential in zebrafish models [8], meriting further scrutiny. Finally, the interaction of Ayurveda medicines with conventional drugs is also of clinical and translational interest. For example, Indian ginseng potentiates antiepileptic activity of diazepam [90], gugul has synergistic effect with hypolipidemics in humans, whereas tulsi is hepatotoxic when co-administered with paracetamol in rats [91]. Given the value of zebrafish in drug screening, especially HTS [92], the use of such sensitive models to probe CNS interactions between Ayurvedic medicines, and/or their interaction with other (e.g., allopathic) drugs, becomes critically important.
5. Future directions
While there is a growing support for assaying CNS effects of various Ayurvedic medicines in zebrafish, their underlying molecular mechanisms remain largely understudied, complicating phenotypical translation and their comparison with other drugs, in terms of both efficiency and targets. It is especially important given multiple Ayurvedic plants with potential CNS effects, and the fact that many representatives of different plant families exert similar (e.g., promnestic and anxiolytic) profiles in clinically or animal models (Table 1, 1-Supplementary material, and 2). Together, this necessitates further studies into potential shared neurobehavioral domains targeted by Ayurvedic medicines across taxa. For example, nootropic and anxiolytic CNS effects in animal models are prevalent among Ayurvedic medicines (Fig. 2), correlating with their commonly reported antioxidant, anti-inflammatory and neuroprotective activity in vivo (Tables 1 and 1-Supplementary material). Taken together, this raises the possibility that behavioral and molecular effects of Ayurvedic medicines may be interrelated. Hence, this not only suggests some shared molecular pathways underlying beneficial CNS effects of Ayurvedic medicine, but also implies that cognitive/behavioral responses may be due to improved neuroprotection and anti-oxidant effects.
Fig. 2.
Summary of common behavioral and physiological CNS effects involved in preclinical activity of Ayurvedic medicines. Data represent the occurrence of specific effects in plans discussed here and listed in Table 1-Supplementary material. GABA - gamma aminobutyric acid.
Moreover, only relatively few Ayurvedic plants have been tested for CNS research in zebrafish models presently (Table 2), resulting in a lack of critical mass of empirical evidence in the field. The use of Ayurvedic plants extract in different sexes, strains and ages of zebrafish is also a challenge since different fish may respond differently to anxiolytic compounds and drugs that affect their general locomotor activity. With the growing number of Ayurvedic plants with well-established CNS effects (Table 1-Supplementary material), further research is needed to fill this knowledge gap utilizing zebrafish models.
6. Potential challenges and limitations
In general, treating brain disorders is complicated by low drug bioavailability and ineffective delivery, unclear genetic risk factors, often unsuitable model organisms for drug discovery and failure to use individual (‘personalized’) medicine-based approaches. As various animal models become used for screening Ayurvedic drugs (Table 1, Table 2), there are also some challenges and limitations in this field, including problematic unavailability of standards for raw materials, identification of bioactive compounds and pharmacognostic analyses, low bioequivalence and safety concerns. The dose standardization for bioactive molecules from Ayurvedic plants is also an important, yet often neglected consideration, especially since animal doses often differ from traditional doses recommended clinically.
Potential adverse effects of Ayurvedic herbal plants also present a concern, especially since few studies have explored them in humans and animals. For example, water hyssop causes poor fertility in rats [93], but not in humans, whereas Indian pennywort may cause severe neurological problems at high doses, including headache, nausea, dizziness and extreme drowsiness [58]. Thus, clinical trials are needed to confirm biosafety profile of Ayurvedic plants, as well as their comparative studies with allopathic medicines, to evaluate their efficacy and safety profiles for the use in humans. Furthermore, herbal constituents undergo in vivo biotransformation, which may lower the efficacy of Ayurvedic plants, and can differ between fish, rodents and humans. There are also difficulties in assessing active molecules in pharmacokinetic studies [94], especially since the crude raw herbal material may differ for geographical locations, climatic conditions, harvesting methods and/or collection protocols, hereby complicating standardizing the drug treatment.
7. Conclusion
Ayurvedic medicines represent affordable and effective folk medicines that may lead to identifying novel drug targets and the discovery of structurally and functionally novel drugs for human brain disorders. The use of zebrafish models is growing rapidly in CNS drug discovery [51], including Ayurvedic medicine (Table 2), albeit not without translational and practical challenges discussed above. Multiple open questions also remain in this field (Table 3), warranting further studies. For example, to what extent zebrafish physiology recapitulates clinical therapy? Are there reliable, evolutionarily conserved biomarkers relevant to therapeutic potential of Ayurvedic medicines that are shared between fish and humans? Does Ayurvedic therapy exert longer-term genomic, epigenetic or epigenomic effects in zebrafish brain? And, finally, can modern nanotechnologies enhance the efficacy of Ayurvedic medicines in zebrafish CNS models? For instance, nanomedicine is increasingly used in drug development, reducing damage to healthy cells and optimizing the consumption of drug dosage [95]. Thus, incorporating nanotechnology in traditional Ayurveda may eliminate one of the major pitfalls of metal toxicity by converting treatments into biological nanoparticles, as in SB and other bhasmas - Ayurvedic preparations using incinerated metals [96]. Testing this and other Ayurvedic medicines in zebrafish will support the growing utility of these fish in probing Ayurvedic drug therapy and studying potential molecular mechanisms of its CNS effects.
Table 3.
Selected open questions related the use of Ayurvedic plants in zebrafish model.
Questions |
---|
|
|
CRediT authorship contribution statement
Pallab Chakraborty: Conceptualization, Writing – original draft, Writing – review & editing. Subharthi Pal: Conceptualization, Writing – original draft, Writing – review & editing. David S. Galstyan: Writing – original draft, Writing – review & editing. Elena V. Petersen: Writing – original draft, Writing – review & editing. Tamara G. Amstislavskaya: Writing – original draft, Writing – review & editing. Tatiana O. Kolesnikova: Writing – original draft, Writing – review & editing. Murilo S. de Abreu: Conceptualization, Writing – original draft, Writing – review & editing. Allan V. Kalueff: Conceptualization, Writing – original draft, Writing – review & editing, Funding acquisition, Supervision.
Declaration of generative AI in scientific writing
None of such technologies were utilized during the preparation of this manuscript.
Funding sources
A.V.K. is supported by the School of Science and Suzhou Key Municipal Laboratory of Neurobiology and Cell Signaling of Xi'an Jiaotong-Liverpool University (Suzhou, China). The funders had no role in the design, analyses and interpretation of the submitted study, or decision to publish.
Conflict of interest
Authors declare no conflicts of interest.
Acknowledgements
MSA (FAPERGS) research Research Support Foundation of the State of Rio Grande do Sul fellowships 24/2551-0001333-4.
Footnotes
Supplementary data to this article can be found online at https://doi.org/10.1016/j.jaim.2025.101169.
Appendix A. Supplementary data
The following is the Supplementary data to this article:
References
- 1.Martins P. The history of traditional indian medicine from beggining to present day. Int J Adv Res. 2018;6:1195–1201. [Google Scholar]
- 2.Rao R.V., et al. Ayurvedic medicinal plants for Alzheimer's disease: a review. Alzheimers Res Ther. 2012;4(3):1–9. doi: 10.1186/alzrt125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Reddy P.H., Manczak M., Yin X., Grady M.C., Mitchell A., Tonk S.…Kumar S. Protective effects of Indian spice curcumin against amyloid-β in Alzheimer's disease. J Alzheim Dis. 2018;61(3):843–866. doi: 10.3233/JAD-170512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kennedy D.O., Wightman E.L. Herbal extracts and phytochemicals: plant secondary metabolites and the enhancement of human brain function. Adv Nutr. 2011;2(1):32–50. doi: 10.3945/an.110.000117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Dubey T., Chinnathambi S. Brahmi (Bacopa monnieri): an ayurvedic herb against the Alzheimer's disease. Arch Biochem Biophys. 2019;676 doi: 10.1016/j.abb.2019.108153. [DOI] [PubMed] [Google Scholar]
- 6.Obulesu M., Rao D.M. Effect of plant extracts on Alzheimer's disease: an insight into therapeutic avenues. J Neurosci Rural Pract. 2011;2(1):56–61. doi: 10.4103/0976-3147.80102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hankey A. A test of the systems analysis underlying the scientific theory of Ayurveda's Tridosha. J Alternative Compl Med. 2005;11(3):385–390. doi: 10.1089/acm.2005.11.385. [DOI] [PubMed] [Google Scholar]
- 8.Biswas S., Balodia N., Bellare J. Comparative neurotoxicity study of mercury-based inorganic compounds including Ayurvedic medicines Rasasindura and Kajjali in zebrafish model. Neurotoxicol Teratol. 2018;66:25–34. doi: 10.1016/j.ntt.2018.01.007. [DOI] [PubMed] [Google Scholar]
- 9.Biswas S., Bellare J. Ayurvedic processing of α-HgS gives novel physicochemistry and distinct toxicokinetics in zebrafish. Chemosphere. 2020;251 doi: 10.1016/j.chemosphere.2020.126295. [DOI] [PubMed] [Google Scholar]
- 10.Ruhela R.K., Soni S., Medhi B. Therapeutic potential of Withania somnifera in CNS disorders: a neuropharmacological review. Eur J Med Plants. 2016:1–12. [Google Scholar]
- 11.Dar N.J. Neurodegenerative diseases and Withania somnifera (L.): an update. J Ethnopharmacol. 2020 doi: 10.1016/j.jep.2020.112769. [DOI] [PubMed] [Google Scholar]
- 12.Prakash J., et al. Neuroprotective role of Withania somnifera root extract in Maneb–Paraquat induced mouse model of parkinsonism. Neurochem Res. 2013;38(5):972–980. doi: 10.1007/s11064-013-1005-4. [DOI] [PubMed] [Google Scholar]
- 13.Sehgal N., et al. Withania somnifera reverses Alzheimer's disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci USA. 2012;109(9):3510–3515. doi: 10.1073/pnas.1112209109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kulkarni S., Dhir A. Withania somnifera: an Indian ginseng. Progress in neuro-psychopharmacol. Biol. Psychiatr. 2008;32(5):1093–1105. doi: 10.1016/j.pnpbp.2007.09.011. [DOI] [PubMed] [Google Scholar]
- 15.Kumar P., Kumar A. Possible neuroprotective effect of Withania somnifera root extract against 3-nitropropionic acid-induced behavioral, biochemical, and mitochondrial dysfunction in an animal model of Huntington's disease. J Med Food. 2009;12(3):591–600. doi: 10.1089/jmf.2008.0028. [DOI] [PubMed] [Google Scholar]
- 16.Kaurav B.P., et al. Influence of Withania somnifera on obsessive compulsive disorder in mice. Asian Pac J Tropical Med. 2012;5(5):380–384. doi: 10.1016/S1995-7645(12)60063-7. [DOI] [PubMed] [Google Scholar]
- 17.Raghav S., et al. Randomized controlled trial of standardized Bacopa monniera extract in age-associated memory impairment. Indian J Psychiatry. 2006;48(4):238–242. doi: 10.4103/0019-5545.31555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Abdul Manap A.S., et al. Bacopa monnieri, a neuroprotective lead in alzheimer disease: a review on its properties, mechanisms of action, and preclinical and clinical studies. Drug Target Insights. 2019;13 doi: 10.1177/1177392819866412. 1177392819866412-1177392819866412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Konar A., Gautam A., Thakur M.K. Bacopa monniera (CDRI-08) upregulates the expression of neuronal and glial plasticity markers in the brain of scopolamine induced amnesic mice. Evid base Compl Alternative Med : eCAM. 2015;2015:837012. doi: 10.1155/2015/837012. 837012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Debnath T., Kim D.H., Lim B.O. Natural products as a source of anti-inflammatory agents associated with inflammatory bowel disease. Molecules. 2013;18(6):7253–7270. doi: 10.3390/molecules18067253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chaudhari K.S., et al. Neurocognitive effect of nootropic drug Brahmi (Bacopa monnieri) in Alzheimer's disease. Ann Neurosci. 2017;24(2):111–122. doi: 10.1159/000475900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Singh B., et al. Neuroprotective effects of Bacopa monnieri in Parkinson's disease model. Metab Brain Dis. 2020;35(3):517–525. doi: 10.1007/s11011-019-00526-w. [DOI] [PubMed] [Google Scholar]
- 23.Kulkarni S., Dhir A. An overview of curcumin in neurological disorders. Indian J Pharmaceut Sci. 2010;72(2):149. doi: 10.4103/0250-474X.65012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kulkarni S.K., Bhutani M.K., Bishnoi M. Antidepressant activity of curcumin: involvement of serotonin and dopamine system. Psychopharmacology. 2008;201(3):435. doi: 10.1007/s00213-008-1300-y. [DOI] [PubMed] [Google Scholar]
- 25.Sharma S., Chopra K., Kulkarni S.K. Effect of insulin and its combination with resveratrol or curcumin in attenuation of diabetic neuropathic pain: participation of nitric oxide and TNF‐alpha. Phytother Res. 2007;21(3):278–283. doi: 10.1002/ptr.2070. [DOI] [PubMed] [Google Scholar]
- 26.Sharma R., et al. Traditional Ayurvedic and herbal remedies for Alzheimer's disease: from bench to bedside. Expert Rev Neurother. 2019;19(5):359–374. doi: 10.1080/14737175.2019.1596803. [DOI] [PubMed] [Google Scholar]
- 27.Di Pierro Francesco R.G., et al. Comparative evaluation of the pain-relieving properties of a lecithinized formulation of curcumin (Meriva®), nimesulide, and acetaminophen. J Pain Res. 2013;6:201. doi: 10.2147/JPR.S42184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Khotimah H., et al. Standardized centella asiatica increased brain-derived neurotrophic factor and decreased apoptosis of dopaminergic neuron in rotenone-induced zebrafish. GSTF J Psychol. 2015;1:22–27. [Google Scholar]
- 29.Vaidya A.D., Devasagayam T.P. Recent advances in Indian herbal drug research guest editor: thomas Paul asir devasagayam current status of herbal drugs in India: an overview. J Clin Biochem Nutr. 2007;41(1):1–11. doi: 10.3164/jcbn.2007001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rao S.B., Chetana M., Uma Devi P. Centella asiatica treatment during postnatal period enhances learning and memory in mice. Physiol Behav. 2005;86(4):449–457. doi: 10.1016/j.physbeh.2005.07.019. [DOI] [PubMed] [Google Scholar]
- 31.Veerendra Kumar M.H., Gupta Y.K. Effect of different extracts of Centella asiatica on cognition and markers of oxidative stress in rats. J Ethnopharmacol. 2002;79(2):253–260. doi: 10.1016/s0378-8741(01)00394-4. [DOI] [PubMed] [Google Scholar]
- 32.Soumyanath A., et al. Centella asiatica extract improves behavioral deficits in a mouse model of Alzheimer's disease: investigation of a possible mechanism of action. Int J Alzheimers Dis. 2012;2012 doi: 10.1155/2012/381974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gupta Y., Kumar M.V., Srivastava A. Effect of Centella asiatica on pentylenetetrazole-induced kindling, cognition and oxidative stress in rats. Pharmacol Biochem Behav. 2003;74(3):579–585. doi: 10.1016/s0091-3057(02)01044-4. [DOI] [PubMed] [Google Scholar]
- 34.Joshi H., Parle M. Nardostachys jatamansi improves learning and memory in mice. J Med Food. 2006;9(1):113–118. doi: 10.1089/jmf.2006.9.113. [DOI] [PubMed] [Google Scholar]
- 35.Oguis G.K., et al. Butterfly pea (Clitoria ternatea), a cyclotide-bearing plant with applications in agriculture and medicine. Front Plant Sci. 2019;10:645. doi: 10.3389/fpls.2019.00645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Jamshidi N., Cohen M.M. The clinical efficacy and safety of Tulsi in humans: a systematic review of the literature. Evid base Compl Alternative Med. 2017;2017 doi: 10.1155/2017/9217567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Saxena R.C., et al. Efficacy of an extract of ocimum tenuiflorum (OciBest) in the management of general stress: a Double-blind, Placebo-controlled Study. Evid base Compl Alternative Med. 2012;2012 doi: 10.1155/2012/894509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bennadi S.J., Krishna K. Protection of zonisamide induced memory impairment by tulsi extract and piracetam on mice. Int. J. Health Allied Sci. 2014;3(3):170. [Google Scholar]
- 39.Mohan L., Amberkar M.V., Kumari M. Ocimum sanctum linn.(TULSI)-an overview. Int J Pharmaceut Sci Rev Res. 2011;7(1):51–53. [Google Scholar]
- 40.Raghavendra M., et al. Role of Ocimum sanctum in the experimental model of Alzheimer's disease in rats. Int J Green Pharm. 2009;3(1) [Google Scholar]
- 41.Butler M.I., et al. The gut microbiome in social anxiety disorder: evidence of altered composition and function. Transl Psychiatry. 2023;13(1):95. doi: 10.1038/s41398-023-02325-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Barberio B., et al. Prevalence of symptoms of anxiety and depression in patients with inflammatory bowel disease: a systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2021;6(5):359–370. doi: 10.1016/S2468-1253(21)00014-5. [DOI] [PubMed] [Google Scholar]
- 43.Hall C.V., et al. Brain signatures of chronic gut inflammation. Front Psychiatr. 2023;14 doi: 10.3389/fpsyt.2023.1250268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lontai L., et al. Burden of mental health among patients with inflammatory bowel disease—a cross-sectional study from a tertiary IBD center in Hungary. J Clin Med. 2024;13(7):2002. doi: 10.3390/jcm13072002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Steer E. A cross comparison between Ayurvedic etiology of Major Depressive Disorder and bidirectional effect of gut dysregulation. J Ayurveda Integr Med. 2019;10(1):59–66. doi: 10.1016/j.jaim.2017.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lopresti A.L., et al. Efficacy of a curcumin extract (Curcugen™) on gastrointestinal symptoms and intestinal microbiota in adults with self-reported digestive complaints: a randomised, double-blind, placebo-controlled study. BMC Complement Med Ther. 2021;21(1):40. doi: 10.1186/s12906-021-03220-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Choudhary D., Bhattacharyya S., Bose S. Efficacy and safety of ashwagandha (Withania somnifera (L.) dunal) root extract in improving memory and cognitive functions. J Diet Suppl. 2017;14(6):599–612. doi: 10.1080/19390211.2017.1284970. [DOI] [PubMed] [Google Scholar]
- 48.Bhattacharya S.K., et al. Anxiolytic-antidepressant activity of Withania somnifera glycowithanolides: an experimental study. Phytomedicine. 2000;7(6):463–469. doi: 10.1016/S0944-7113(00)80030-6. [DOI] [PubMed] [Google Scholar]
- 49.Hawiset T., et al. Anxiolytic and antidepressant-like activities of aqueous extract of Azadirachta indica A. Juss. flower in the stressed rats. Heliyon. 2022;8(2) doi: 10.1016/j.heliyon.2022.e08881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Faucher P., et al. Synergistic effects of Lacticaseibacillus rhamnosus GG, glutamine, and curcumin on chronic unpredictable mild stress-induced depression in a mouse model. Benef Microbes. 2022;13(3):253–264. doi: 10.3920/BM2021.0188. [DOI] [PubMed] [Google Scholar]
- 51.Patton E.E., Zon L.I., Langenau D.M. Zebrafish disease models in drug discovery: from preclinical modelling to clinical trials. Nat Rev Drug Discov. 2021:1–18. doi: 10.1038/s41573-021-00210-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Stewart A.M., et al. Zebrafish models for translational neuroscience research: from tank to bedside. Trends Neurosci. 2014;37(5):264–278. doi: 10.1016/j.tins.2014.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kalueff A.V., Stewart A.M., Gerlai R. Zebrafish as an emerging model for studying complex brain disorders. Trends Pharmacol Sci. 2014;35(2):63–75. doi: 10.1016/j.tips.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Stewart A.M., Gerlai R., Kalueff A.V. Developing highER-throughput zebrafish screens for in-vivo CNS drug discovery. Front Behav Neurosci. 2015;9:14. doi: 10.3389/fnbeh.2015.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nasiadka A., Clark M.D. Zebrafish breeding in the laboratory environment. ILAR J. 2012;53(2):161–168. doi: 10.1093/ilar.53.2.161. [DOI] [PubMed] [Google Scholar]
- 56.Zon L.I., Peterson R.T. In vivo drug discovery in the zebrafish. Nat Rev Drug Discov. 2005;4(1):35–44. doi: 10.1038/nrd1606. [DOI] [PubMed] [Google Scholar]
- 57.Rajini P.S. Ayurvedic herbal preparations in neurological disorders. Elsevier; 2023. Utilization of zebrafish as a preclinical model for obtaining insights into molecular mechanisms of the neuroprotective role of ayurvedic herbal preparations; pp. 521–540. [Google Scholar]
- 58.Gohil K.J., Patel J.A., Gajjar A.K. Pharmacological review on Centella asiatica: a potential herbal cure-all. Indian J Pharmaceut Sci. 2010;72(5):546. doi: 10.4103/0250-474X.78519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Xu C.-L., et al. Asiaticoside: attenuation of neurotoxicity induced by MPTP in a rat model of Parkinsonism via maintaining redox balance and up-regulating the ratio of Bcl-2/Bax. Pharmacol Biochem Behav. 2012;100(3):413–418. doi: 10.1016/j.pbb.2011.09.014. [DOI] [PubMed] [Google Scholar]
- 60.Mohanty R., et al. Withania somnifera leaf extract ameliorates benzo [a] pyrene-induced behavioral and neuromorphological alterations by improving brain antioxidant status in zebrafish (Danio rerio) Zebrafish. 2016;13(3):188–196. doi: 10.1089/zeb.2015.1215. [DOI] [PubMed] [Google Scholar]
- 61.Khatri D.K., Juvekar A.R. Abrogation of locomotor impairment in a rotenone-induced Drosophila melanogaster and zebrafish model of Parkinson's disease by ellagic acid and curcumin. Int J Nutrition Pharmacol Neurol Dis. 2016;6(2):90. [Google Scholar]
- 62.Abidar S., et al. The aqueous extract from Ceratonia siliqua leaves protects against 6-hydroxydopamine in zebrafish: understanding the underlying mechanism. Antioxidants. 2020;9(4):304. doi: 10.3390/antiox9040304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Maddula K., Kumar V.P., Anusha J. Assessment of aqueous extract of ocimum sanctum leaves in memory enhancement and preventing memory impairment activities in zebra fish model. J Basic Clin Pharm. 2017;8(3) [Google Scholar]
- 64.Serikuly N., et al. Effects of acute and chronic arecoline in adult zebrafish: anxiolytic-like activity, elevated brain monoamines and the potential role of microglia. Prog Neuro Psychopharmacol Biol Psychiatr. 2020 doi: 10.1016/j.pnpbp.2020.109977. [DOI] [PubMed] [Google Scholar]
- 65.Ngadni M.A., et al. Clitorienolactones and isoflavonoids of clitorea ternatea roots alleviate stress-like symptoms in a reserpine-induced zebrafish model. Molecules. 2021;26(14) doi: 10.3390/molecules26144137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mudgal P., et al. Assessment of anxiolytic activity of brahmi (bacopa monnieri) in zebrafish model system. J Nat Remedies. 2023:661–670. [Google Scholar]
- 67.de Abreu M.S., et al. Modeling gut-brain interactions in zebrafish. Brain Res Bull. 2019 doi: 10.1016/j.brainresbull.2019.03.003. [DOI] [PubMed] [Google Scholar]
- 68.Kishore N., et al. Implications of Asparagus racemosus and Terminalia chebula extracts on oxazolone induced inflammatory bowel disease in Danio rerio (zebrafish) Biocatal Agric Biotechnol. 2023;51 [Google Scholar]
- 69.Bajaj S., Vohora S.B. Anti-cataleptic, anti-anxiety and anti-depressant activity of gold preparations used in Indian system of medicine. Indian J Pharmacol. 2000;32(6):339–346. [Google Scholar]
- 70.Biswas S., et al. Neuroprotective effects of nanogold-based Ayurveda medicine Suvarna Bhasma against rotenone-induced Parkinson's-like model. J Ayurveda Integr Med. 2024;15(1) doi: 10.1016/j.jaim.2023.100854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Biswas S., et al. Physicochemical characterization of Suvarna Bhasma, its toxicity profiling in rat and behavioural assessment in zebrafish model. J Ethnopharmacol. 2020;249 doi: 10.1016/j.jep.2019.112388. [DOI] [PubMed] [Google Scholar]
- 72.Alli R.A. What are the treatments for autism? 2020. https://www.webmd.com/brain/autism/understanding-autism-treatment WebMD:[Available from:
- 73.Ruknuddin G., et al. Anti-inflammatory and analgesic activities of Dashanga Ghana: an Ayurvedic compound formulation. Int J Nutrition Pharmacol Neurol Dis. 2013;3(3):303–308. [Google Scholar]
- 74.Agarwal V., Abhijnhan A., Raviraj P. Ayurvedic medicine for schizophrenia. Cochrane Database Syst Rev. 2007;(4) doi: 10.1002/14651858.CD006867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sarkar S., et al. Add-on effect of Brahmi in the management of schizophrenia. J Ayurveda Integr Med. 2012;3(4):223. doi: 10.4103/0975-9476.104448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Demin K.A., et al. Developing zebrafish experimental animal models relevant to schizophrenia. Neurosci Biobehav Rev. 2019;105:126–133. doi: 10.1016/j.neubiorev.2019.07.017. [DOI] [PubMed] [Google Scholar]
- 77.Elakkiya V., et al. Advances in Ayurvedic medicinal plants and nanocarriers for arthritis treatment and management: a review. J Herb Med. 2020;24 [Google Scholar]
- 78.Ranade S.Y., Gaud R.S. Current strategies in herbal drug delivery for arthritis: an overview. Int J Pharmaceut Sci Res. 2013;4(10):3782. [Google Scholar]
- 79.Zhang Z., Huang G. Micro-and nano-carrier mediated intra-articular drug delivery systems for the treatment of osteoarthritis. J Nanotechnol. 2012:2012. [Google Scholar]
- 80.Aguiar G.P.S., et al. Micronization of N-acetylcysteine by supercritical fluid: evaluation of in vitro and in vivo biological activity. J Supercrit Fluids. 2017;130:282–291. [Google Scholar]
- 81.Carrilho E., Tavares M.C.H., Lanças F.M. Fluidos supercríticos em química analítica. II. Cromatografia com fluido supercrítico: instrumentação. Quim Nova. 2003;26(5):687–693. [Google Scholar]
- 82.Decui L., et al. Micronized resveratrol shows promising effects in a seizure model in zebrafish and signalizes an important advance in epilepsy treatment. Epilepsy Res. 2020;159 doi: 10.1016/j.eplepsyres.2019.106243. [DOI] [PubMed] [Google Scholar]
- 83.Genario R., et al. Sex differences in behavior and neuropharmacology of zebrafish. Eur J Neurosci. 2019;0(0) doi: 10.1111/ejn.14438. [DOI] [PubMed] [Google Scholar]
- 84.Cremer J.E. In: Alterations of chemical equilibrium in the nervous system. Lajtha A., editor. Springer US; Boston, MA: 1971. Body temperature and drug effects; pp. 311–323. [Google Scholar]
- 85.Reardon S. Sex matters in experiments on party drug—in mice. Nature News. 2017 [Google Scholar]
- 86.de Abreu M.S., Kalueff A.V. Of mice and zebrafish: the impact of the experimenter identity on animal behavior. Lab Anim. 2021;50(1):7. doi: 10.1038/s41684-020-00685-9. 7. [DOI] [PubMed] [Google Scholar]
- 87.Biswas S., Bellare J. Explaining Ayurvedic preparation of Rasasindura, its toxicological effects on NIH3T3 cell line and zebrafish larvae. J Ayurveda Integr Med. 2022;13(2) doi: 10.1016/j.jaim.2021.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Best J., Alderton W.K. Zebrafish: an in vivo model for the study of neurological diseases. Neuropsychiatric Dis Treat. 2008;4(3):567. doi: 10.2147/ndt.s2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Challal S., et al. Zebrafish bioassay-guided microfractionation for the rapid in vivo identification of pharmacologically active natural products. CHIMIA Int J Chem. 2012;66(4):229–232. doi: 10.2533/chimia.2012.229. [DOI] [PubMed] [Google Scholar]
- 90.Joshi K., et al. Studies of ashwagandha (Withania somnifera dunal) Int J Pharmaceut Biol Archiv. 2016;7:1–11. [Google Scholar]
- 91.Dhanya R., Shukla A.K. Drug interactions between ayurvedic and allopathic medicines–A review. World J Pharmaceut Res. 2017;6:478–486. [Google Scholar]
- 92.Zhang T., Peterson R.T. In: The zebrafish in biomedical research. Cartner S.C., et al., editors. Academic Press; 2020. Chapter 51 - zebrafish as a platform for drug screening; pp. 659–675. [Google Scholar]
- 93.Singh A., Singh S.K. Evaluation of antifertility potential of Brahmi in male mouse. Contraception. 2009;79(1):71–79. doi: 10.1016/j.contraception.2008.07.023. [DOI] [PubMed] [Google Scholar]
- 94.Sharma A.K., et al. Problems associated with clinical trials of Ayurvedic medicines. Revista Brasileira de Farmacognosia. 2010;20:276–281. [Google Scholar]
- 95.Pelaz B., et al. Diverse applications of nanomedicine. ACS Nano. 2017;11(3):2313–2381. doi: 10.1021/acsnano.6b06040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Roopashree S., Anitha J., Rashmi S. Advent of nanomaterial in modern health science and ayurveda. Mater Today Proc. 2021;46:9096–9101. [Google Scholar]
- 97.Amin H., et al. Nootropic (medhya) effect of Bhāvita Śaṇkhapuṣpī tablets: a clinical appraisal. Ancient Sci Life. 2014;34(2):109. doi: 10.4103/0257-7941.153476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Amin H., et al. Shankhapushpi (Convolvulus pluricaulis Choisy): validation of the Ayurvedic therapeutic claims through contemporary studies. Int J Green Pharm. 2014;8(4) [Google Scholar]
- 99.Lyle N., et al. The role of antioxidant properties of Nardostachys jatamansi in alleviation of the symptoms of the chronic fatigue syndrome. Behav Brain Res. 2009;202(2):285–290. doi: 10.1016/j.bbr.2009.04.005. [DOI] [PubMed] [Google Scholar]
- 100.Karkada G., et al. Nardostachys jatamansi extract prevents chronic restraint stress-induced learning and memory deficits in a radial arm maze task. J Nat Sci Biol Med. 2012;3(2):125. doi: 10.4103/0976-9668.101879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kumar M.V., Gupta Y. Effect of different extracts of Centella asiatica on cognition and markers of oxidative stress in rats. J Ethnopharmacol. 2002;79(2):253–260. doi: 10.1016/s0378-8741(01)00394-4. [DOI] [PubMed] [Google Scholar]
- 102.Saxena G., et al. Gugulipid, an extract of Commiphora whighitii with lipid-lowering properties, has protective effects against streptozotocin-induced memory deficits in mice. Pharmacol Biochem Behav. 2007;86(4):797–805. doi: 10.1016/j.pbb.2007.03.010. [DOI] [PubMed] [Google Scholar]
- 103.Husnul Khotimah M.A., Sutiman Bambang Sumitro, Aris Widod Mochamad. Decreasing a-synuclein aggregation by methanolic extract of Centella asiatica in zbrafish Parkinson's model. Asian Pac J Trop Biomed. 2015;5(11):948–954. [Google Scholar]
- 104.Bernardi M., Dias S., Barbosa V. Neurotoxicity of neem commercial formulation (Azadirachta indica A. Juss) in adult zebrafish (Danio rerio) Environ Toxicol Pharmacol. 2013;36(3):1276–1282. doi: 10.1016/j.etap.2013.10.002. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.