Skip to main content

REVIEW article

Front. Nutr., 18 August 2022
Sec. Food Chemistry
Volume 9 - 2022 | https://doi.org/10.3389/fnut.2022.972379

Pharmacological attributes of Bacopa monnieri extract: Current updates and clinical manifestation

  • 1Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
  • 2School of Biotechnology, IFTM University, Moradabad, India
  • 3Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt
  • 4Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah, Saudi Arabia
  • 5Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
  • 6Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia

Bacopa monnieri has been used for centuries in Ayurvedic medicine, alone or in combination with other herbs, as a memory and learning enhancer, sedative, and anti-epileptic. This review aimed to highlight the health benefits of B. monnieri extracts (BME), focusing on anti-cancer and neurodegenerative diseases. We examined the clinical studies on phytochemistry and pharmacological application of BME. We further highlighted the mechanism of action of these extracts in varying types of cancer and their therapeutic implications. In addition, we investigated the underlying molecular mechanism in therapeutic interventions, toxicities, safety concerns and synergistic potential in cognition and neuroprotection. Overall, this review provides deeper insights into the therapeutic implications of Brahmi as a lead formulation for treating neurological disorders and exerting cognitive-enhancing effects.

Introduction

Bacopa monnieri (Brahmi) is a well-known perennial, creeping herb possessing bioactive formulation in the Indian Ayurveda system, implicated in the therapeutic management of numerous diseases. This herb was used by Ancient Vedic scholars due to its pharmacological effect, especially as a nerve tonic and nootropic booster. However, to better understand Bacopa monnieri's role in several neurological disorders and memory-related diseases, it is necessary to understand its active phytochemical constituents and underlying mechanism of action. Bioactive components of Brahmi belong to alkaloids, saponins, flavonoids, triterpenes and cucurbitacin, having potential role in neuroprotection (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1. An overview of the pharmacological effects of major bioactive constituents of Bacopa monnieri.

Among various medicinal Ayurvedic herbs, Bacopa monnieri is considered a herb of grace and commonly known as Brahmi, belonging to the family Scrophulariaceae. It is a small creeping herb with numerous branches, small oblong leaves, and light purple flowers found throughout the Indian subcontinent in wet, damp, and marshy area (1). Brahmi has had a rich historical and religious background for more than 1,400 years. It has been reported for various pharmacological activities. Brahmi is used as a brain booster by amassing the information evolved through experience over years and years because it acts as a rejuvenator for the brain and nervous system.

The extracts of B. monnieri are well-recognized for their antioxidant activity with numerous modes of action to protect the brain against oxidative damage and cognitive decline in the elderly. The cognition-promoting roles of B monnieri can be due to the antioxidant effects of alcoholic extracts and bacoside (2). Based on animal study results, the B. monnieri extract and bacosides were shown to enhance antioxidant status in the brain region of the hippocampus, frontal cortex, and striatum (3). In the diabetic model, it ameliorates diabetes-induced-oxidative stress (4). The evidence suggests that in vivo chronic cigarette smoke exposure enhances oxidative stress, and bacoside A was found to protect against cigarette smoking-induced cerebrovascular diseases by decreasing the formation of free radicals through its antioxidant potential (5).

Earlier research also reported a dose-dependent free radical scavenging ability and a protective effect of methanolic extract B. monnieri against DNA (6, 7). In vitro and in vivo studies in C. elegans done by Phulara et al. (8) provide evidence that B monnieri has antioxidant activity and is capable of up-regulating the expression of the gene hsp-16.2 associated with stress tolerance, which greatly improves the lifetime of C. elegans under stress conditions (8).

The nephroprotective efficacy of B. monnieri in mice against Tracolimus-induced kidney toxicity has been reported (9). This protective efficacy is accompanied by a significant attenuation of oxidative stress and maybe through free radical scavenging activity of B. monnieri. This evidence suggests that B. monnieri might have potential as an adjunct therapy in which free radical production plays a vital role and would be useful in advancing novel B. monnieri herbal drugs for various stress-related human complications.

The extract of Brahmi and its isolated valuable therapeutic agents have been extensively investigated for their nootropic effects, antioxidant, antimicrobial properties and analgesic activity, etc. These traditional pharmacological claims have been bolstered by large-scale research and clinical studies (10). Brahmi has been the focus of research as a versatile therapeutic agent for various disorders and neurodegenerative diseases.

This review article addresses the major phytochemical profile and pharmacological attributes emphasizing the neuroprotective role of Bacopa monnieri. We further highlighted the underlying biochemical mechanisms of action of Bacopa monnieri in neuroprotection and other diseases.

Bioactive constituents and their functional significance

Bacopa monnieri plant is rich in clinically critical secondary metabolites such as saponins, alcohols, steroids, alkaloids, glycosides, sterol glycosides, phenylethanoid glycosides, sugars, amino acids, flavonoids and cucurbitacins (1113). In addition, Brahmin, Hydrocotyline, Nicotine, Herpestine, D-mannitol, stigmasterol, glutamic acid, aspartic acid, alanine, and serine are specific amino acids are present in the extracts of Bacopa monnieri. Structural features of different components of Bacopa monnieri extracts are illustrated in Figure 2. The major component are saponins comprising bacosides, bacopasides (13), Bacosaponins (14, 15), Betulinic acid, etc.

FIGURE 2
www.frontiersin.org

Figure 2. Representative structures of major phytoconstituents present in extracts of Bacopa monnieri. Structures were downloaded from PubChem (https://pubchem.ncbi.nlm.nih.gov) with their corresponding PubChem CIDs, Bacoside A (92043183), Bacoside B (121596009), Bacogenin A (101600046), Bacopaside I (21599442), Bacopasaponin C (21599443), Cucurbitacin B (5281316), Bacosine (71312547), and Stigmasterol (5280794).

Bacosides are significant components of Bacopa monnieri and play essential roles in neuronal health. Structurally, bacoside-A (PubChem ID: 92043183) is an amphiphilic chemical compound containing both sterol and sugar moieties. Deepak et al. (16) identified and characterized 12 analogs of the bacosides, known as bacopasides I-XII. Most of the glycosides have sugar chains attached to the C-3 only (classified as monodesmosides) and in few to both C-3 and C-20 (classified as bidesmosides) of the aglycone unit (17). They protected the cytotoxicity and DNA damage of neurons implicated in Alzheimer's disease (AD) and repaired the impaired neurons by enhancing kinase activity and neuronal synthesis (18). Bacosides A and B are responsible for most neuropharmacological and nootropic effects (14, 16). Bacoside A contains four saponin glycosides viz. bacopaside II, bacopaside X, bacoside A3 and bacopasaponin C (16). In contrast, bacoside B only varies in optical rotation with bacoside A and consists of bacopaside IV, V, N1, & N2 (16, 19, 20). Bacoside A is pharmacologically more active than bacoside B.

Bacogenin A1–A5 is the acid hydrolyzed derivatives of bacosides (2123), and among which ebelin lactone (bacogenin A4) (24) is the major component (25). Ramasamy and co-workers suggested that ebelin lactone and bacogenin A1 bind highly to CNS receptors. Bacopasides I-XII are important saponins that interact with sterols and are involved in membrane disruption (13, 2628).

Other jujubogenin and pseudojujubogenin derivatives whose role is yet to be explored are termed Bacopasaponin A-H (29, 30). Among these, bacopasaponin C comprises 0.3–0.6% of the ethanolic extracts of BM (31). It is a glycosidic pseudojujobogenin with glucose and rhamnose as sugar units (28). This terminal glucose moiety is self-targeting toward cells with their specific receptors and is responsible for the antileishmanial property of bacopasaponin C (32).

Cucurbitacin displays varied types of biological properties in plants and animals. Four cucurbitacins, bacitracin A-D and cucurbitacin E, isolated from the ethanolic extract of the dichloromethane (DCM) portion of the BM plant (33). Betulinic acid and its derivative dihydro betulinic acid (IC50 = 0.5 μM) are the most potent pentacyclic triterpenoid inhibitor of eukaryotic topoisomerase I for anti-cancer drug designing (34). In addition, bacosine protected against oxidative damage in alloxanized diabetes and increased peripheral glucose consumption. Bacosine administration also upturned weight loss in diabetic rats and prohibited in vitro glycosylation of hemoglobin (35).

Cancer prevention

B. monnieri has an anti-cancer efficacy on different types of cancer. Palethorpe et al. (36) reported that bacopaside I and bacopaside II, a terpenoid from B. monnieri, can synergistically block the functional activity of the membrane transport system aquaporin, AQP1 is also reported to contribute to tumor progression. The reduced transcriptional expression of AQP1 inhibits proliferation, migration and invasion in breast cancer cell lines.

Similarly, Pei et al. (37) reported that bacopaside I and bacopaside II blocked AQP1 and inhibited colon cancer cell lines (37). This work is further supported by Smith et al. (38). Bacopaside II has been shown to activate autophagy by inhibiting G2/M cell cycle transition and inducing apoptosis of low and high AQP1-expressing colon cancer cells.

Based on these findings, bacopasides have been proposed as potential novel lead compounds for the pharmaceutical production of selective AQP blockers in cancer treatment. Hepatocellular carcinoma (HCC) represents the fifth most common cancer globally and is related to mortality worldwide (39). B. monnieri's alcohol extract has been highlighted as an effective antioxidant, free radical scavenger, and a potential anti-lipid peroxidative agent (3, 4042).

Janani et al. (43) showed that BM extract, Bacoside A, can prevent N-nitrosodiethylamine (DEN)-induced hepatoma by inhibiting lipid peroxidation and by enhancing the levels of antioxidant enzymes in Wistar albino rat. In another study, Janani et al. (author?) (44) investigated the effect of Bacoside A on the activities and expression of matrix metalloproteases (MMPs) enzymes, i.e., MMP-2 and MMP-9 in DEN-induced HCC. They reported that Bacoside A employs its anti-metastatic effect against DEN-induced HCC by suppressing the activities and expressions of MMP-2 and MMP-9 enzymes responsible for metastasis in various tumors (44).

Nitrobenzene is a hazardous air pollutant and is considered a human carcinogen that affects the liver, brain, blood, and stomach. The ethanolic extract of BM at the dose of 200 mg/kg showed a good hepatoprotective effect in nitrobenzene-induced liver damage in mice model by an increase in SOD, CAT and GPx enzymes and by normalizing the serum marker enzymes such as aspartate transaminase, alanine transaminase, and alkaline phosphatase. In contrast, the levels of these serum marker enzymes increased in the carcinogen-administered mice models (45).

Another common and aggressive tumor that causes the highest deaths worldwide is glioblastoma (GBM). A glioblastoma is a brain tumor with an inferior prognosis that is highly vascularized, infiltrative, and exacerbates its tumor potential. All these features are therapeutic objectives in glioblastoma treatment, including surgical removal accompanied by chemotherapy and radiotherapy (46, 47). Existing therapies have not adequately handled the patient, so classical therapies have had to expand and integrate new alternative approaches, like natural compounds. Other targets in the treatment of glioblastoma are the inhibition of the notch signaling pathway that contributes to a decrease in glioblastoma cell proliferation and self-renewal (48, 49), the receptor for an epidermal growth factor (EGFR) (50), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) (51). Natural substances are emerging as potential therapies to address GBM growth (5255). A previous study also documented the apoptotic activity of bacoside A in brain tumor cells, GBM (56).

Neuroblastoma is an embryonic cancer of the autonomic nervous system arising from the embryonic sympathoadrenal lineage of the neural crest. For children between 1 and 5 years of age, neuroblastoma is the main cause of death from pediatric cancer and accounts for around 13% of pediatric cancer mortality (57). The neuroprotective potential of B. monnieri has been studied with its active ingredients, such as Bacopasaponins, Betulinic acid, Bacoside A and B, etc. Thus, researchers have taken advantage of the neuroprotective property of B. monnieri in searching for natural remedies for this pediatric cancer.

Several studies reported that the extract of this herbal plant prevents hydrogen peroxide-induced oxidative damage in human neuroblastoma cell lines (58, 59). These findings suggested that B. monnieri effectively treats different forms of cancer and can shield against brain damage, and improve brain development.

Management of diabetes nephropathy

Diabetes mellitus (DM) is a chronic metabolic condition with life-threatening complications characterized by hyperglycemia, hyperlipidemia, hyper aminoacidemia, and hyperinsulinemia. According to the seventh edition of the World Diabetes Atlas released by the International Diabetes Federation (IDF), as of 2015, about 415 million people worldwide live with diabetes. This number will likely increase to 642 million by 2040 (60). Drugs are available to monitor and treat diabetic patients, but complete diabetes recovery has not been reported.

Alternative to these drugs, plants provide potential antidiabetic effects and are commonly used in many conventional medicine schemes to prevent diabetes (6164). In a study conducted by Gosh et al., ethanolic extract of the aerial parts of B. monnieri was evaluated against antioxidant and antihyperglycemic activity in the Wistar mice model and elucidated that BME prevents significant elevation of glycosylated hemoglobin in vitro with IC50 value being 11.25 μg/ml that is comparable with the control drug α-tocopherol (65). This IC50 value reached 7.44 μg/mL when treated with only Bacosine, a triterpene from B. monnieri (35).

The previous study has also indicated that an isolate of BM, stigmasterol is effective in streptozotocin-nicotinamide-induced diabetic nephropathy (DN), i.e., it inhibits the progression of chronic complications of diabetes via reducing the formation of advanced glycation end products and amelioration of oxidative stress (66). Moreover, the previous research demonstrated that B. monnieri reduced serum glucose and increased diabetic rat body weight (4, 67).

Parkinson's disease

Parkinson's disease (PD) is a slowly progressive, degenerative disorder characterized by degeneration of nerve cells in the substantia nigra region and aggregation of a key protein, alpha-synuclein, in the striatum and adjacent brain regions. B. monnieri modulates PD (68, 69). Evidence from the animal model showed the anti-Parkinson's effect of B. monnieri extract. Bacosides, an alcoholic extract of B. monnieri, was explored in a Caenorhabditis elegans model where it exhibited decreased aggregation of α-synuclein and prevented dopaminergic neurodegeneration, restoring nematode lipid material (70). Another group of researchers reported that Bacopa treatment to the MPTPP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced parkinsonian mice model offers nigrostriatal dopaminergic neuroprotection against MPTP-induced parkinsonism by modulating the behavioral effects of oxidative stress and apoptotic machinery (71).

Moreover, the effect of the extract of B. monnieri leaf was studied on transgenic model flies of Drosophila that expressed normal human alpha-synuclein in their neurons. The extract improves behavioral defects and decreases oxidative stress and apoptosis in the flies of PD model brains (72). All these findings confirm the efficacy of B. monnieri as a novel therapeutic for PD treatment.

Alzheimer's disease

A significant and growing public health concern is AD. It is associated with cognitive impairment and dementia and is characterized by the accumulation of amyloid-β peptides in senile plaques and abnormally phosphorylated tau proteins (7375). The root cause of a group of neurodegenerative disorders collectively known as “tauopathies” was confirmed to be hyperphosphorylation of tau protein. The detrimental effect is the loss of affinity between this protein and the microtubules, increased fibrillary aggregate production, and accumulated insoluble neurofibrillary tangles (7678).

In animal models of AD, Bacopa has been shown to suppress beta-amyloid deposits in the brain (79). This herb showed a significant memory-enhancing influence. Research demonstrated that this herb facilitates acquisition, retention, and recovery (80). Evidence also showed that many herbs effective against AD (8185). Brahmi and its active component have exclusively been explored in AD treatment among all herbs. Recently, it has been reported that alcoholic extract of B. monnieri significantly improves escape latency time in the Morris water maze test and facilitates the reduction of neurons and cholinergic neuron densities in a rat model of AD (86). A detailed mechanism of action of B. monnieri in neurodegenerative disease is described in Figure 3.

FIGURE 3
www.frontiersin.org

Figure 3. The action mechanism of Bacopa monnieri against neurodegenerative diseases.

Holcomb et al. (87) studied the PSAPP rodent model. They revealed that the administration of Bacopa extract to mice expressing APP and PSEN-1 mutation diminished amyloidogenic proteins Aβ40 and Aβ42 levels in the brain by ~60%. In vitro study explored by Mathew et al. (88) on anti-amyloidogenic potential found that Bacopa almost completely reduced the development of amyloid fibrils and greatly separated the preformed amyloid fibrils.

An exciting in silico study showed that Bacopasaponin G and Bacopasaponin N2, two saponins from B. monnieri, might be effective in AD therapy. Compared with Donepezil, these two saponins exert a more favorable binding affinity with the Caspase-3 and tau-protein kinase I (TPK I) receptors, therapeutic targets in AD (89). These findings indicate that this herb is a potential cognitive enhancer and promises to be a novel agent in AD.

Pre-clinical studies

Preclinical testing was done to validate the effectiveness of B. monnieri extract as a herbal medicinal drug. According to several in vitro and in vivo investigations, BME appears to be helpful in the treatment/prevention of neurodegenerative diseases and other age-related disorders.

In vitro studies

Limpeanchob et al. (90) evaluated the neuroprotective effect of B. monnieri extracts by assessing the viability of cultures of primary cortical neuron cells treated with 50 μM aggregated Aβ 25–35 in the presence and absence of BME. In the presence of 100 μg/ml of BME, the survival rate of the cultured cells increases while the survival rate reduces in the absence of BME.

Neuronal death induced by amyloid peptide exhibited a high 2-fold rise in acetylcholine esterase (AChE) concentration, while those treated with Brahmi extract had a near-normal concentration of AChE. This study validates that Brahmi extract increases the neuronal survival rate of neuronal cells by suppressing AChE activity. Likewise, BME pre-treatment significantly reduced scopolamine-induced PC12 cell death, and viability was restored at 85.75% of control with 100 g/mL extract of BM. Pre-treatment with BME reduced the release of lactate dehydrogenase (LDH) by up to 22.42% of the total, compared to 30% in the scopolamine-treated group. BME also improved scopolamine effects by decreasing AChE and increasing muscarinic-1 receptor and BDNF expression (91).

Another study conducted by Malishev et al. (92) in the SH-SY5Y cell line demonstrated that Bacoside A at 50 M significantly inhibited cytotoxicity, fibrillation, and, in particular, membrane interactions of A (1–42) (A42). Bhatia et al. (93) evaluated the protective effect of BME against hydrogen peroxide (H2O2) induced oxidative damage in a cellular model of neuroblastoma IMR32 cells. These protective effects possibly were associated with an increase in glutathione levels, enhancing endogenous defense machinery and maintaining membrane integrity. To better characterize the neuroprotective effect of BME at the molecular level, RT-PCR and immunofluorescence were used to observe the expression of NF200 (an intermediate filament in neurons) and heat shock proteins (HSP70 and mortalin). In normal conditions, NF200, HSP70 and mortalin are assigned for cytoarchitecture and axonal transport, proper functioning of the cell under normal and stress conditions and cell proliferation, respectively. The elevated expression of these stress markers because of H2O2-induced oxidative stress causes brain injury, and varied pathological conditions, including cerebral ischemia and neurodegenerative diseases. The expression of these three oxidative stress markers was significantly alleviated after the pre-treatment of BME, which supports the neuroprotective effect of BME.

Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor, with a high proliferative rate and invasiveness. Notch1 signaling has been associated with anti-apoptotic behavior in various cellular contexts. Notch1 receptor promotes glioblastoma cells' survival by regulating the anti-apoptotic Mcl-1 protein. Inhibition of Notch1 signaling through knockdown of notch receptors sensitizes glioblastoma cells for anti-tumor treatment. The notch signaling pathway has thus proved to be a novel therapeutic target in treating GBM (94). Introduction of Bacoside A in human glioblastoma cell line U-87 MG causes cell cycle arrest and apoptosis by inhibiting the Notch1 receptors and sensitizes glioblastoma cells to apoptosis (95).

In vivo studies

B. monnieri as a neuroprotective agent was evaluated by in vivo studies using various experimental models. In vivo study demonstrated in Mus musculus (house mouse) that D-Galactose and Sodium nitrite induced impaired cognitive functions that were significantly ameliorated by administering BME (100 mg/kg of body weight), confirming that BME has anti-Alzheimer's properties (96). The cholinergic and glutamatergic networks and their interactions are involved in cognitive dysfunction associated with AD. The glutamatergic system is essential in regulating synaptic plasticity and cognition (97, 98).

Administration of B. monnieri ameliorates olfactory bulbectomy (OBX) induced cognition dysfunction in mouse models by the protection of cholinergic systems and by activating the synaptic proteins to induce synaptic plasticity (99). Administration of BME was found to facilitate the scopolamine effect by downregulating cholinesterase (ChE) in albino mice which was observed by improved performance on the Morris Water Maze Test. In this study, BME treatment showed a significant increase in step-down latency (SDL), which may prove to be a reliable memory restorative agent in curing dementia seen in AD (100). Gamma-aminobutyric acid (GABA) and brain-derived neurotrophic factor (BDNF) are well-known to be related to synapse recovery, neuronal survival, and neuronal protection. Thus, GABAergic transmission is thought to play a role in neurodegenerative diseases' etiology and regenerative processes by maintaining equivalent neurotransmission in the CNS.

Therefore, GABAergic transmission stability may be a therapeutic solution in many neurodegenerative disorders. Piyabhan et al. (101) demonstrated the effects of Brahmi in a PCP-induced schizophrenic-like model, including partial restoration of cognitive deficit and neuroprotection. They elucidated its underlying mechanism of action by increasing GABAergic neurons. Singh et al. (102) investigated the effect of BME on MPTP-induced nigrostriatal dopaminergic neurodegeneration in mice.

Their study demonstrated that BME has neuroprotective and neurorescuive effects. The overall in vitro and in vivo studies suggested that BME can mitigate memory impairment and neurodegenerative disorders. Table 1 outlines the specific impact of B. monnieri extract on various study designs (in vitro and in vivo) of neurodegenerative diseases.

TABLE 1
www.frontiersin.org

Table 1. Summary of in vitro and in vivo studies.

B. monnieri extracts in clinical trials

Manifold clinical studies provide evidence in the form of placebo-controlled, randomized, and double-blind trials to support the cognitive benefits of B. monnieri supplementation. The potential of this ayurvedic medicine as a dietary supplement for prevention or as a candidate drug to cure acute and chronic neurodegenerative conditions seems relevant. Accordingly, various clinical trials evaluated their efficacy in mental aging neuropsychiatric diseases.

To investigate the effects of BME (KeenMind) on cognitive performance in healthy adults with an age spectrum between 18 and 60 years, Stough et al. (103) used a double-blind placebo-controlled trial and a series of well-validated neuropsychological assessments. A total of 46 participants were randomly allocated to 1 of 2 treatment conditions. Capsules of 320 mg of BME were given for 12 weeks which significantly improved verbal learning, early information processing, and memory strengthening in participants compared to non-treated groups. The authors support the previously published studies that chronic dosages of B. monnieri (KeenMind) for 90 days improve accuracy in more complex cognitive tasks (104). A significant difference in the finding of the later study from those of the earlier study (103) was the lack of reduction in state anxiety and involvement of speeded computerized tasks in the present study. Also, Roodenrys et al. (105) studied the effects of 90 days of BM (KeenMind) supplementation on memory-enhancing out-turn in 76 volunteers with an age spectrum between 40 and 65 years.

Another randomized, placebo-controlled, double-blind clinical study was performed by Morgan and Stevens (106) to see the effect of Brahmi (BacoMind) in improving older adults' memory performance. They all used the same dosage amount to see the cognitive impact of BME (KeenMind). Their findings revealed that Brahmi (BacoMind) could be used as a memory enhancer. Peth Nui et al. (107) had evaluated the effect of 300 mg of Brahmi on cognitive processing, attention, working memory, cholinergic and monoaminergic functions in 60 healthy adults. Another group of researchers, in their double-blind, placebo-controlled clinical trial of 320 mg and 640 mg doses involving 17 healthy adults (aged between 18 and 44 years), reported the acute effects of Brahmi (CDRI 08) on stress and mood swings caused by multitasking (108).

In another study, the randomized, double-blind placebo-controlled efficacy of B. monnieri extract (CDRI 08) enhanced cognitive performances. Here, 100 subjects aged between 6 and 14 were given 1 × 160 mg capsule of either Bacopa or placebo if weight is between 20 and 35 kg or 2 × 160 mg capsules of either Bacopa or placebo per day weight is above 35 kg for 16 weeks. This study demonstrated that BM is significantly beneficial for the symptoms of hyperactivity or attention deficit hyperactivity disorder (ADHD) and suggestive of cognitive improvement (109). Anhedonia (the reduced ability to experience pleasure) is a hallmark symptom of various neuropsychiatric diseases that leads to poor mental health outcomes throughout one's life and predicts poor psychosocial functioning.

Moreover, another research utilized a randomized, double-blind, placebo-controlled study involving 60 medical students with already high cognitive functions, which showed that B. monnerri extract (Bacognize) (150 mg twice a day) given for 6 weeks significantly improved cognitive function along with a significant rise in serum calcium (p < 0.05) (still within standard range i.e., 9–11 mg/dL). The cognitive performance was validated by examining various neuropsychological tests (logical memory test, digit span memory task, etc.). These detailed memory assessments will provide a better insight into subtle memory deficits. Their study finding revealed no change in the brain's attention and sensory-motor performance, indicating that BME reduces participants' distractibility but somewhat improves cognitive functions (110).

A pilot study was performed to assess the Brahmi as a memory enhancer and its safety and tolerability in elderly patients of either sex. Each individual received 250 mg of Brahmi tablet (b.i.d.) for 3 months. All patients showed a rise in cognitive fitness with no significant adverse effect (111). In a recent study, it was found that the administration of B. monnieri extract (300 mg bid) in 19 patients for 4 weeks proved effective in the treatment of anhedonia when compared with the controls (23 patients) who have treated with citalopram 40 mg (TAU) (112). Table 2 summarizes the stated clinical trials of BME in humans.

TABLE 2
www.frontiersin.org

Table 2. Summary of various clinical studies.

Toxicity and safety concerns

Despite the increased demand for herbal formulations in the market, there are several issues related to their safety. The safety of herbal medicine stands at a high-water mark with a significant increase in global consumption. There is currently confusion and prejudice regarding the safety of the herbal medication. As a result, public awareness, objective comprehension, and neutral and fair interpretation are required. Moreover, caregivers must understand what the drug is, its use, and how it should be administered. Medications should be kept out of the reach of people with cognitive impairment. It is one of the most demanding tasks for scientists and researchers to investigate the efficacy, adverse effects, and serious contaminants from mixtures of herbal formulations. The most important reasons for herbal drug toxicity are improper identification of plant material, contamination of herbs with toxins, pesticides, and heavy metals, and their interaction with conventional drugs upon concomitant intake. If strict standardization and quality control parameters have not been followed, the errors, including contamination of heavy metals, and excessive alcohol generation in formulations, lead to reverse effects (113).

However, the herbal formulation can boost pharmaceuticals' effect and reduce the dosage of susceptible individuals. Even the most potent poison can become the best drug (114). Thus, there should be more focus on improving these herbs' bioavailability with the generation of minimum side effects. Quality control should be applied throughout the various processing stages, from the raw material to the finished product. A flow chart given in Figure 4 shows the standardized protocol for developing herbal formulations.

FIGURE 4
www.frontiersin.org

Figure 4. The Global concept of standardization, quality evaluation and pharmacological validation for the development of herbal medicine.

Several studies have evaluated the effectiveness of BME in an intoxicated animal model. In Sodium fluoride (NaF) intoxicated Swiss albino female mice, 300 mg/kg of BME reversed the effects of fluoride and impeded neuropathological alterations by restoring the cholinergic system and by decreasing the oxidative stress (115).

In 30% alcohol plus carbon tetrachloride (CCl4), intoxication of the Wistar Albino rats leads to hepatic oxidative stress that has been reversed by administering 100 mg and 200 mg/kg body weight/day of 70% ethanol extract of BM (116). Some studies revealed that 40 mg/kg of mBME can manage morphine-related hepatotoxicity and nephrotoxicity (117, 118).

Paraquat (PQ) exposure causes increased oxidative stress and mitochondrial dysfunction, followed by apoptosis and cell death. A standardized extract of B. monnieri neutralizes the PQ-mediated toxicity in Drosophila by optimizing oxidative stress, restoring ATP levels and decreasing apoptosis via inhibition of active JNK and cleaved Caspase-3 (119). Heavy metal toxicities have been recognized as a major public health risk. It interferes with the functions of various organs (liver, kidney) and systems (CNS, hematopoietic system).

Heavy metals cause oxidative stress and enzyme inhibition by interacting with the function of essential cations. Their accumulation ultimately leads to intellectual and behavioral impairments. Several studies in an in vivo model revealed the mitigating effect of BME on lead and aluminum-induced oxidative stress (120, 121). The methylated form of mercury (Hg), known as methylmercury (MeHg), is a ubiquitous environmental pollutant. MeHg-exposed rodents undergo oxidative stress in the cerebellum region leading to a deficit in motor performance.

The administration of Brahmi at 250 mg/mL concentration ameliorates the MeHg-Induced oxidative impairments (122). Consequently, BME has been proven promising in traditional medicine to protect the brain from oxidative damage resulting from heavy metal toxicity. The transcription factor nuclear factor-2 erythroid-related factor-2 (Nrf2) regulates the production of antioxidant genes via endogenous antioxidant (GSH level) mechanisms while also acting anti-inflammatory. Okadaic acid (OKA) administration resulted in memory impairment, decreased Nrf2 levels and caused oxidative stress and neuroinflammation in rats. Oral administration of BM (40 and 80 mg/kg) and melatonin (20 mg/kg) restored Nrf2 levels, decreased oxidative stress, and strengthened endogenous antioxidants. Thus, Nrf2 level modulation improved rats' spatial learning in the Morris water maze (123). B. monnieri was also found to attenuate trimethyltin (TMT)-induced cognitive impairments in mice by protecting the cholinergic system that promotes neurodegeneration in the dentate gyrus regions and protects the hippocampal neuron (124).

A study was conducted using wild type C. elegans model to evaluate the effect of BM and hexane extract (but not the ethanol extract) on glutamate exposure-induced AD parameters like mitochondrial stress and ROS production, as well as to assess the effects of hexane extract on the aging and life span of the model organism. Administration of 10 mg/ml B.monnieri hexane extract using ANOVA followed by Dunnett's post-hoc test showed a reduction in mitochondrial stress (P < 0.05) and ROS production (P < 0.0001) in cultured neuronal cells. Also, B. monnieri hexane extract at a dose of 7 and 10 mg/ml could extend the median and maximum lifespan and reduce the effects of aging in aged worms, thus, proving that BM hexane extract can be a potential prophylaxis agent against oxidative and mitochondrial damage and can be used as a therapeutic agent in aged patients (125).

Inappropriately, B. monnieri extracts did not induce toxicity or other symptoms in both female and male acute and chronic oral toxicity testing (126). Based on the results described above, we can conclude that B. monnieri is non-toxic and have the efficiency to reverse the adverse effect caused by toxic agents. Hence, B. monnieri is reliably safe for use for pharmacological purposes. However, more in-depth analyses are still required to explore the toxicity of the herb for human health-promoting benefits. The cited toxicity and prevention findings are summarized in Table 3.

TABLE 3
www.frontiersin.org

Table 3. Efficacy of B. monnieri to attenuate the adverse effect caused by various toxic agents.

Conclusion and future prospects

Bacopa monnieri extracts are extensively used to enhance memory and intelligence in Ayurvedic and Unani medicine systems. Extracts isolated from B.monnieri such as flavonoids, saponins, and triterpenes prevent oxidative and mitochondrial/ER stress and increase the aging duration in C. elegans. The present review summarizes recent findings on the potential health benefits of B. monnieri. Extracts of Bacopa monnieri such as Bacoside A, Bacoside B, Bacosaponins, and Betulinic acid play significant role in neuroprotection. The neuroprotective properties of these bioactive components include reduction of ROS, neuroinflammation, aggregation inhibition of amyloid-β and improvement of cognitive and learning behavior. Major phytoconstituents of B. monnieri are saponins such as bacoside A3, bacopaside II, X and bacopasaponin C and its isomer. Several authors reported inhibitory effects of bacoside on the glioma cell's viability and proliferation, indicating promising anti-cancer activity for the treatment of glioblastoma. Finally, we conclude that B. monnieri extracts could be implicated in treating Alzheimer's disease and other neurological disorders. However, future investigations are required to compare the neuroprotective effect of B. monnieri extracts with standard drugs to establish systematic clinical uses.

Author contributions

Conceptualization and writing—review and editing: UF and MH. Methodology: UF and SR. Software and resources: SAh and SAl. Validation: WE, IK, and RA. Formal analysis: MA. Investigation: AI. Data curation: SR. Writing—original draft preparation: UF. Visualization: SAh. Supervision: SAl and IK. Project administration: WE. Funding acquisition: MH. All authors have read and agreed to the published version of the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the National Medicinal Plants Board (NMPB) and Ministry of AYUSH, Government of India [Z. 18017/187/CSS/R&D/DL-01/2019-20-NMPB-IVA].

Acknowledgments

UF thank the Indian Council of Medical Research-Department of Health Research (ICMR-DHR) for the award of Young Scientist. MH thank the Council of Scientific and Industrial Research (Grant No. 27(0368)/20/EMRII). The authors sincerely thank the Department of Science and Technology, Government of India, for the FIST support (FIST program No. SR/FST/LSII/2020/782).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Jain PK, Das D, Jain P, Jain P, Jain PK. Pharmacognostic and pharmacological aspect of Bacopa monnieri - a review. Innovare J Ayurvedic Sci. (2016) 4, 796-11.

Google Scholar

2. Chaudhari KS, Tiwari NR, Tiwari RR, Sharma RS. Neurocognitive effect of nootropic drug Brahmi (Bacopa monnieri) in Alzheimer's disease. Ann Neurosci. (2017) 24:111–22. doi: 10.1159/000475900

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Bhattacharya S, Bhattacharya A, Kumar A, Ghosal S. Antioxidant activity ofBacopa monniera in rat frontal cortex, striatum and hippocampus. Phytother Res. (2000) 14:174–9. doi: 10.1002/(SICI)1099-1573(200005)14:3andlt;174::AID-PTR624andgt;3.0.CO;2-O

CrossRef Full Text | Google Scholar

4. Kapoor R, Srivastava S, Kakkar P. Bacopa monnieri modulates antioxidant responses in brain and kidney of diabetic rats. Environ Toxicol Pharmacol. (2009) 27:62–9. doi: 10.1016/j.etap.2008.08.007

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Anbarasi K, Vani G, Balakrishna K, Devi CS. Effect of bacoside A on brain antioxidant status in cigarette smoke exposed rats. Life Sci. (2006) 78:1378–84. doi: 10.1016/j.lfs.2005.07.030

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Anand T, Naika M, Swamy M, Khanum F. Antioxidant and DNA damage preventive properties of Bacopa monniera (L.) wettst. Free Rad Antioxid. (2011) 1:84–90. doi: 10.5530/ax.2011.1.13

CrossRef Full Text | Google Scholar

7. Russo A, Izzo AA, Borrelli F, Renis M, Vanella A. Free radical scavenging capacity and protective effect of Bacopa monniera L. on DNA damage. Phytother Res. (2003) 17:870–5. doi: 10.1002/ptr.1061

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Phulara SC, Shukla V, Tiwari S, Pandey R. Bacopa monnieri promotes longevity in Caenorhabditis elegans under stress conditions. Pharmacogn Mag. (2015) 11:410. doi: 10.4103/0973-1296.153097

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Oyouni AAA, Saggu S, Tousson E, Mohan A, Farasani A. Mitochondrial nephrotoxicity induced by tacrolimus (FK-506) and modulatory effects of Bacopa monnieri (Farafakh) of Tabuk region. Pharmacognosy Res. (2019) 11. doi: 10.4103/pr.pr_100_18

CrossRef Full Text | Google Scholar

10. Abdul Manap AS, Vijayabalan S, Madhavan P, Chia YY, Arya A, Wong EH, 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) 13:1177392819866412. doi: 10.1177/1177392819866412

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Bhandari P, Kumar N, Singh B, Kaul VK. Cucurbitacins from Bacopa monnieri. Phytochemistry. (2007) 68:1248–54. doi: 10.1016/j.phytochem.2007.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Chakravarty AK, Sarkar T, Nakane T, Kawahara N, Masuda K. New phenylethanoid glycosides from Bacopa monniera. Chem Pharm Bull. (2002) 50:1616–8. doi: 10.1248/cpb.50.1616

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Rauf K, Subhan F, Al-Othman A, Khan I, Zarrelli A, Shah M. Preclinical profile of bacopasides from Bacopa monnieri (BM) as an emerging class of therapeutics for management of chronic pains. Curr Med Chem. (2013) 20:1028–37. doi: 10.2174/092986713805288897

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Chatterji N, Rastogi R, Dhar M. Chemical examination of Bacopa monniera Wettst: Part I-Isolation of chemical constituents. Indian J Chem. (1963) 1:212–5.

Google Scholar

15. Mathew J, Paul J, Nandhu M, Paulose C. Bacopa monnieri and Bacoside-A for ameliorating epilepsy associated behavioral deficits. Fitoterapia. (2010) 81:315–22. doi: 10.1016/j.fitote.2009.11.005

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Deepak M, Amit A. The need for establishing identities of'bacoside A and B', the putative major bioactive saponins of Indian medicinal plant Bacopa monnieri. Phytomedicine. (2004) 11:264. doi: 10.1078/0944-7113-00351

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Bhandari P, Sendri N, Devidas SB. Dammarane triterpenoid glycosides in Bacopa monnieri: a review on chemical diversity and bioactivity. Phytochemistry. (2020) 172:112276. doi: 10.1016/j.phytochem.2020.112276

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Kishore K, Singh M. Effect of bacosides, alcoholic extract of Bacopa monniera Linn. (brahmi), on experimental amnesia in mice. Indian J Exp Biol. (2005) 43:640–5.

PubMed Abstract | Google Scholar

19. Deepak M, Amit A. ‘Bacoside B’—the need remains for establishing identity. Fitoterapia. (2013) 87:7–10. doi: 10.1016/j.fitote.2013.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Sivaramakrishna C, Rao CV, Trimurtulu G, Vanisree M, Subbaraju GV. Triterpenoid glycosides from Bacopa monnieri. Phytochemistry. (2005) 66:2719–28. doi: 10.1016/j.phytochem.2005.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chandel R, Kulshreshtha D, Rastogi R. Bacogenin-A3: a new sapogenin from Bacopa monniera. Phytochemistry. (1977) 16:141–3. doi: 10.1016/0031-9422(77)83039-2

CrossRef Full Text | Google Scholar

22. Kulshreshtha D, Rastogi R. Bacogenin-A1: a novel dammarane triterpene sapogenin from Bacopa monniera. Phytochemistry. (1973) 12:887–92. doi: 10.1016/0031-9422(73)80697-1

CrossRef Full Text | Google Scholar

23. Kulshreshtha D, Rastogi R. Bacogenin A2: a new sapogenin from bacosides. Phytochemistry. (1974) 13:1205–6. doi: 10.1016/0031-9422(74)80101-9

CrossRef Full Text | Google Scholar

24. Kulshreshtha D, Rastogi R. Identification of ebelin lactone from Bacoside A and the nature of its genuine sapogenin. Phytochemistry. (1973) 12:2074–6. doi: 10.1016/S0031-9422(00)91552-8

CrossRef Full Text | Google Scholar

25. Rastogi S, Pal R, Kulshreshtha DK. Bacoside A3? A triterpenoid saponin from Bacopa monniera. Phytochemistry. (1994) 36:133–7. doi: 10.1016/S0031-9422(00)97026-2

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Chakravarty AK, Garai S, Masuda K, Nakane T, Kawahara N. Bacopasides III—V: three new triterpenoid glycosides from Bacopa monniera. Chem Pharma Bull. (2003) 51:215–7. doi: 10.1248/cpb.51.215

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Chakravarty AK, Sarkar T, Masuda K, Shiojima K, Nakane T, Kawahara N. Bacopaside I and II: two pseudojujubogenin glycosides from Bacopa monniera. Phytochemistry. (2001) 58:553–6. doi: 10.1016/S0031-9422(01)00275-8

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Garai S, Mahato SB, Ohtani K, Yamasaki K. Dammarane-type triterpenoid saponins from Bacopa monniera. Phytochemistry. (1996) 42:815–20. doi: 10.1016/0031-9422(95)00936-1

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Garai S, Mahato SB, Ohtani K, Yamasaki K. Bacopasaponin DA pseudojujubogenin glycoside from Bacopa monniera. Phytochemistry. (1996) 43:447–9. doi: 10.1016/0031-9422(96)00250-6

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Mahato SB, Garai S, Chakravarty AK. Bacopasaponins E and F: two jujubogenin bisdesmosides from Bacopa monniera. Phytochemistry. (2000) 53:711–4. doi: 10.1016/S0031-9422(99)00384-2

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Huangteerakul C, Aung HM, Thosapornvichai T, Duangkaew M, Jensen AN, Sukrong S, et al. Chemical-genetic interactions of Bacopa monnieri constituents in cells deficient for the DNA repair endonuclease RAD1 appear linked to vacuolar disruption. Molecules. (2021) 26:1207. doi: 10.3390/molecules26051207

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Sinha J, Raay B, Das N, Medda S, Garai S, Mahato S, et al. Bacopasaponin C: critical evaluation of anti-leishmanial properties in various delivery modes. Drug Deliv. (2002) 9:55–62. doi: 10.1080/107175402753413181

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Miro M. Cucurbitacins and their pharmacological effects. Phytother Res. (1995) 9:159–68. doi: 10.1002/ptr.2650090302

CrossRef Full Text | Google Scholar

34. Chowdhury AR, Mandal S, Mittra B, Sharma S, Mukhopadhyay S, Majumder HK. Betulinic acid, a potent inhibitor of eukaryotic topoisomerase I: identification of the inhibitory step, the major functional group responsible and development of more potent derivatives. Med Sci Monitor. (2002) 8:BR254–60. doi: 10.12659/MSM.937927

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Ghosh T, Maity TK, Singh J. Antihyperglycemic activity of bacosine, a triterpene from Bacopa monnieri, in alloxan-induced diabetic rats. Planta Med. (2011) 77:804–8. doi: 10.1055/s-0030-1250600

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Palethorpe HM, Smith E, Tomita Y, Nakhjavani M, Yool AJ, Price TJ, et al. Bacopasides I and II act in synergy to inhibit the growth, migration and invasion of breast cancer cell lines. Molecules. (2019) 24:3539. doi: 10.3390/molecules24193539

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Pei JV, Kourghi M, De Ieso ML, Campbell EM, Dorward HS, Hardingham JE, et al. Differential inhibition of water and ion channel activities of mammalian aquaporin-1 by two structurally related bacopaside compounds derived from the medicinal plant bacopa monnieri. Mol Pharmacol. (2016) 90:496–507. doi: 10.1124/mol.116.105882

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Smith E, Palethorpe HM, Tomita Y, Pei JV, Townsend AR, Price TJ, et al. The purified extract from the medicinal plant Bacopa monnieri, bacopaside II, inhibits growth of colon cancer cells in vitro by inducing cell cycle arrest and apoptosis. Cells. (2018) 7:81. doi: 10.3390/cells7070081

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Puoti C. New insights on hepatocellular carcinoma: epidemiology and clinical aspects. Hepatoma Res. (2018) 4:57. doi: 10.20517/2394-5079.2018.67

CrossRef Full Text | Google Scholar

40. Garg A, Kumar A, Nair A, Reddy A. Elemental analysis of brahmi (Bacopa monnieri) extracts by neutron activation and its bioassay for antioxidant, radio protective and anti-lipid peroxidation activity. J Radioanal Nuclear Chem. (2009) 281:53–8. doi: 10.1007/s10967-009-0081-z

CrossRef Full Text | Google Scholar

41. Ghosh T, Maity T, Bose A, Dash GK, Das M. A study on antimicrobial activity of Bacopa monnieri Linn. aerial parts. J Nat Remed. (2006) 6:170–3.

42. Xu Z, Shi M, Tian Y, Zhao P, Niu Y, Liao M. Dirhamnolipid produced by the pathogenic fungus Colletotrichum gloeosporioides BWH-1 and its herbicidal activity. Molecules. (2019) 24:2969. doi: 10.3390/molecules24162969

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Janani P, Sivakumari K, Geetha A, Ravisankar B, Parthasarathy C. Chemopreventive effect of bacoside A on N-nitrosodiethylamine-induced hepatocarcinogenesis in rats. J Cancer Res Clin Oncol. (2010) 136:759–70. doi: 10.1007/s00432-009-0715-0

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Janani P, Sivakumari K, Geetha A, Yuvaraj S, Parthasarathy C. Bacoside A downregulates matrix metalloproteinases 2 and 9 in DEN-induced hepatocellular carcinoma. Cell Biochem Funct. (2010) 28:164–9. doi: 10.1002/cbf.1638

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Menon BR, Rathi M, Thirumoorthi L, Gopalakrishnan V. Potential effect of Bacopa monnieri on nitrobenzene induced liver damage in rats. Indian J Clin Biochem. (2010) 25:401–4. doi: 10.1007/s12291-010-0048-4

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Keime-Guibert F, Chinot O, Taillandier L, Cartalat-Carel S, Frenay M, Kantor G, et al. Radiotherapy for glioblastoma in the elderly. N Engl J Med. (2007) 356:1527–35. doi: 10.1056/NEJMoa065901

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res. (2009) 29:5171–84.

PubMed Abstract | Google Scholar

48. Hovinga KE, Shimizu F, Wang R, Panagiotakos G, Van Der Heijden M, Moayedpardazi H, et al. Inhibition of notch signaling in glioblastoma targets cancer stem cells via an endothelial cell intermediate. Stem Cells. (2010) 28:1019–29. doi: 10.1002/stem.429

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Kanamori M, Kawaguchi T, Nigro JM, Feuerstein BG, Berger MS, Miele L, et al. Contribution of Notch signaling activation to human glioblastoma multiforme. J Neurosurg. (2007) 106:417–27. doi: 10.3171/jns.2007.106.3.417

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Westphal M, Maire CL, Lamszus K. EGFR as a target for glioblastoma treatment: an unfulfilled promise. CNS Drugs. (2017) 31:723–35. doi: 10.1007/s40263-017-0456-6

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Nogueira L, Ruiz-Ontañon P, Vazquez-Barquero A, Moris F, Fernandez-Luna JL. The NFκB pathway: a therapeutic target in glioblastoma. Oncotarget. (2011) 2:646. doi: 10.18632/oncotarget.322

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Abbas M, Kausar S, Cui H. Therapeutic potential of natural products in glioblastoma treatment: targeting key glioblastoma signaling pathways and epigenetic alterations. Clin Transl Oncol. (2020) 22:963–77. doi: 10.1007/s12094-019-02227-3

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Mishra R, Kaur G. Aqueous ethanolic extract of Tinospora cordifolia as a potential candidate for differentiation based therapy of glioblastomas. PLoS ONE. (2013) 8:e78764. doi: 10.1371/journal.pone.0078764

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Racoma IO, Meisen WH, Wang Q-E, Kaur B, Wani AA. Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS ONE. (2013) 8:e72882. doi: 10.1371/journal.pone.0072882

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Tavana E, Mollazadeh H, Mohtashami E, Modaresi SMS, Hosseini A, Sabri H, et al. Quercetin: a promising phytochemical for the treatment of glioblastoma multiforme. BioFactors. (2020) 46:356–66. doi: 10.1002/biof.1605

PubMed Abstract | CrossRef Full Text | Google Scholar

56. John S, Sivakumar K, Mishra R. Bacoside A induces tumor cell death in human glioblastoma cell lines through catastrophic macropinocytosis. Front Mol Neurosci. (2017) 10:171. doi: 10.3389/fnmol.2017.00171

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med. (2015) 66:49–63. doi: 10.1146/annurev-med-011514-023121

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Łojewski M, Pomierny B, Muszyńska B, Krzyzanowska W, Budziszewska B, Szewczyk A. Protective effects of Bacopa monnieri on hydrogen peroxide and staurosporine: induced damage of human neuroblastoma SH-SY5Y cells. Planta Med. (2016) 82:205–10. doi: 10.1055/s-0035-1558166

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Petcharat K, Singh M, Ingkaninan K, Attarat J, Yasothornsrikul S. Bacopa monnieri protects SH-SY5Y cells against tert-Butyl hydroperoxide-induced cell death via the ERK and PI3K pathways. Siriraj Med J. (2015) 67:20–6.

PubMed Abstract | Google Scholar

60. Atlas D. International Diabetes Federation. IDF Diabetes Atlas. 7th ed. Brussels: International Diabetes Federation (2015).

PubMed Abstract | Google Scholar

61. Al-Attar AM, Alsalmi FA. Influence of olive leaves extract on hepatorenal injury in streptozotocin diabetic rats. Saudi J Biol Sci. (2019) 26:1865–74. doi: 10.1016/j.sjbs.2017.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Edwin J, Balakrishnan JS, Chandra JD. Diabetes and herbal medicines. Iran J Pharmacol Therapeut. (2008) 97–106.

63. Kooti W, Farokhipour M, Asadzadeh Z, Ashtary-Larky D, Asadi-Samani M. The role of medicinal plants in the treatment of diabetes: a systematic review. Electron Physician. (2016) 8:1832. doi: 10.19082/1832

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Rao MU, Sreenivasulu M, Chengaiah B, Reddy KJ, Chetty CM. Herbal medicines for diabetes mellitus: a review. Int J PharmTech Res. (2010) 2:1883–92.

Google Scholar

65. Ghosh T, Sengupta P, Dash D, Bose A. Antidiabetic and in vivo antioxidant activity of ethanolic extract of Bacopa monnieri Linn. aerial parts: a possible mechanism of action. Iran J Pharm Res (IJPR). (2008) 7, 61–8.

Google Scholar

66. Kishore L, Kaur N, Singh R. Renoprotective effect of Bacopa monnieri via inhibition of advanced glycation end products and oxidative stress in STZ-nicotinamide-induced diabetic nephropathy. Ren Fail. (2016) 38:1528–44. doi: 10.1080/0886022X.2016.1227920

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Pandey SP, Singh HK, Prasad S. Alterations in hippocampal oxidative stress, expression of AMPA receptor GluR2 subunit and associated spatial memory loss by Bacopa monnieri extract (CDRI-08) in streptozotocin-induced diabetes mellitus type 2 mice. PLoS ONE. (2015) 10:e0131862. doi: 10.1371/journal.pone.0131862

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Hosamani R. The efficacy of Bacopa monnieri extract in modulating Parkinson's disease. In: Genetics, Neurology, Behavior, and Diet in Parkinson's Disease. (2020). Cambridge, MA: Elsevier. p. 609–24. doi: 10.1016/B978-0-12-815950-7.00039-4

CrossRef Full Text | Google Scholar

69. Singh B, Pandey S, Rumman M, Mahdi AA. Neuroprotective effects of Bacopa monnieri in Parkinson's disease model. Metab Brain Dis. (2020) 35:517–25. doi: 10.1007/s11011-019-00526-w

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Jadiya P, Khan A, Sammi SR, Kaur S, Mir SS, Nazir A. Anti-Parkinsonian effects of Bacopa monnieri: insights from transgenic and pharmacological Caenorhabditis elegans models of Parkinson's disease. Biochem Biophys Res Commun. (2011) 413:605–10. doi: 10.1016/j.bbrc.2011.09.010

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Singh B, Pandey S, Yadav SK, Verma R, Singh SP, Mahdi AA. Role of ethanolic extract of Bacopa monnieri against 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced mice model via inhibition of apoptotic pathways of dopaminergic neurons. Brain Res Bull. (2017) 135:120–8. doi: 10.1016/j.brainresbull.2017.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Siddique YH, Mujtaba SF, Faisal M, Jyoti S, Naz F. The effect of Bacopa monnieri leaf extract on dietary supplementation in transgenic Drosophila model of Parkinson's disease. Eur J Integr Med. (2014) 6:571–80. doi: 10.1016/j.eujim.2014.05.007

CrossRef Full Text | Google Scholar

73. Naqvi AAT, Hasan GM, Hassan MI. Targeting tau hyperphosphorylation via kinase inhibition: strategy to address Alzheimer's disease. Curr Top Med Chem. (2020) 20:1059–73. doi: 10.2174/1568026620666200106125910

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Naqvi AAT, Jairajpuri DS, Noman OMA, Hussain A, Islam A, Ahmad F, et al. Evaluation of pyrazolopyrimidine derivatives as microtubule affinity regulating kinase 4 inhibitors: towards therapeutic management of Alzheimer's disease. J Biomol Struct Dyn. (2020) 38:3892–907. doi: 10.1080/07391102.2019.1666745

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Shamsi A, Mohammad T, Anwar S, Alajmi MF, Hussain A, Hassan MI, et al. Probing the interaction of Rivastigmine Tartrate, an important Alzheimer's drug, with serum albumin: attempting treatment of Alzheimer's disease. Int J Biol Macromol. (2020) 148:533–42. doi: 10.1016/j.ijbiomac.2020.01.134

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Bondi MW, Edmonds EC, Salmon DP. Alzheimer's disease: past, present, and future. J Int Neuropsychol Soc. (2017) 23:818–31. doi: 10.1017/S135561771700100X

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Medeiros R, Baglietto-Vargas D, LaFerla FM. The role of tau in Alzheimer's disease and related disorders. CNS Neurosci Ther. (2011) 17:514–24. doi: 10.1111/j.1755-5949.2010.00177.x

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Medina M, Avila J. New perspectives on the role of tau in Alzheimer's disease. Implic Ther Biochem Pharmacol. (2014) 88:540–7. doi: 10.1016/j.bcp.2014.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Dhanasekaran M, Tharakan B, Holcomb LA, Hitt AR, Young KA, Manyam BV. Neuroprotective mechanisms of ayurvedic antidementia botanical Bacopa monniera. Phytother Res. (2007) 21:965–9. doi: 10.1002/ptr.2195

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Ssingh H, Rastogi R, Srimal R, Dhawan B. Effect of bacosides A and B on avoidance response in rats. Phytother Res. (1988) 2:70–5. doi: 10.1002/ptr.2650020205

CrossRef Full Text | Google Scholar

81. Das TK, Hamid M, Das T, Shad KF. Potential of Glyco-withanolides from Withania Somnifera (Ashwagandha) as therapeutic agents for the treatment of Alzheimer's disease. World J Pharm Res. (2015) 4:16–38.

Google Scholar

82. Esfandiari E, Ghanadian M, Rashidi B, Mokhtarian A, Vatankhah AM. The effects of Acorus calamus L. in preventing memory loss, anxiety, and oxidative stress on lipopolysaccharide-induced neuroinflammation rat models. Int J Prev Med. (2018) 9:85. doi: 10.4103/ijpvm.IJPVM_75_18

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Habtemariam S. The therapeutic potential of rosemary (Rosmarinus officinalis) diterpenes for Alzheimer's disease Evid Based Complement Altern Med. (2016) 2016:2680409. doi: 10.1155/2016/2680409

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Kim H-J, Jung S-W, Kim S-Y, Cho I-H, Kim H-C, Rhim H, et al. Panax ginseng as an adjuvant treatment for Alzheimer's disease. J Ginseng Res. (2018) 42:401–11. doi: 10.1016/j.jgr.2017.12.008

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Liao Z, Cheng L, Li X, Zhang M, Wang S, Huo R. Meta-analysis of Ginkgo biloba preparation for the treatment of Alzheimer's disease. Clin Neuropharmacol. (2020) 43:93–9. doi: 10.1097/WNF.0000000000000394

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Uabundit N, Wattanathorn J, Mucimapura S, Ingkaninan K. Cognitive enhancement and neuroprotective effects of Bacopa monnieri in Alzheimer's disease model. J Ethnopharmacol. (2010) 127:26–31. doi: 10.1016/j.jep.2009.09.056

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Holcomb LA, Dhanasekaran M, Hitt AR, Young KA, Riggs M, Manyam BV. Bacopa monniera extract reduces amyloid levels in PSAPP mice. J Alzheimers Dis. (2006) 9:243–51. doi: 10.3233/JAD-2006-9303

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Mathew M, Subramanian S. Evaluation of the anti-amyloidogenic potential of nootropic herbal extracts in vitro. Int J Pharm Sci Res. (2012) 3:4276–80.

Google Scholar

89. Roy S, Chakravarty S, Talukdar P, Talapatra SN. Identification of bioactive compounds present in Bacopa monnieri Linn. against caspase-3 and Tau Protein Kinase I to prevent alzheimer's disease: an in silico study. Pharma Innov. J. (2019) 8:855–61.

Google Scholar

90. 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. J Ethnopharmacol. (2008) 120:112–7. doi: 10.1016/j.jep.2008.07.039

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Pandareesh M, Anand T. Neuromodulatory propensity of Bacopa monniera against scopolamine-induced cytotoxicity in PC12 cells via down-regulation of AChE and up-regulation of BDNF and muscarnic-1 receptor expression. Cell Mol Neurobiol. (2013) 33:875–84. doi: 10.1007/s10571-013-9952-5

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Malishev R, Shaham-Niv S, Nandi S, Kolusheva S, Gazit E, Jelinek R. Bacoside-A, an Indian traditional-medicine substance, inhibits β-amyloid cytotoxicity, fibrillation, and membrane interactions. ACS Chem Neurosci. (2017) 8:884–91. doi: 10.1021/acschemneuro.6b00438

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Bhatia G, Dhuna V, Dhuna K, Kaur M, Singh J. Bacopa monnieri extracts prevent hydrogen peroxide-induced oxidative damage in a cellular model of neuroblastoma IMR32 cells. Chin J Nat Med. (2017) 15:834–46. doi: 10.1016/S1875-5364(18)30017-7

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Fassl A, Tagscherer K, Richter J, Diaz MB, Llaguno SA, Campos B, et al. Notch1 signaling promotes survival of glioblastoma cells via EGFR-mediated induction of anti-apoptotic Mcl-1. Oncogene. (2012) 31:4698–708. doi: 10.1038/onc.2011.615

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Aithal MG, Rajeswari N. Bacoside A induced sub-G0 arrest and early apoptosis in human glioblastoma cell line U-87 MG through notch signaling pathway. Brain Tumor Res Treat. (2019) 7:25–32. doi: 10.14791/btrt.2019.7.e21

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Kunte KB, Kuna Y. Neuroprotective effect of Bacopa monniera on memory deficits and ATPase system in Alzheimer's disease (AD) induced mice. J Sci Innov Res. (2013) 2:719–35.

Google Scholar

97. Cheng Y-J, Lin C-H, Lane H-Y. Involvement of cholinergic, adrenergic, and glutamatergic network modulation with cognitive dysfunction in Alzheimer's disease. Int J Mol Sci. (2021) 22:2283. doi: 10.3390/ijms22052283

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Schaeffer EL, Gattaz WF. Cholinergic and glutamatergic alterations beginning at the early stages of Alzheimer disease: participation of the phospholipase A 2 enzyme. Psychopharmacology. (2008) 198:1–27. doi: 10.1007/s00213-008-1092-0

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Le XT, Pham HTN, Do PT, Fujiwara H, Tanaka K, Li F, et al. Bacopa monnieri ameliorates memory deficits in olfactory bulbectomized mice: possible involvement of glutamatergic and cholinergic systems. Neurochem Res. (2013) 38:2201–15. doi: 10.1007/s11064-013-1129-6

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Kishore D, Babu RS, Begum A, Noor A, Farheen S, Kauser SM, et al. Evaluation of nootropic activity of two marketed drugs of Bacopa monnieri in scopolamine induced amnesic models. Indian J Res Pharmacy Biotechnol. (2018) 6:84–90.

Google Scholar

101. Piyabhan P, Tingpej P, Duansak N. Effect of pre-and post-treatment with Bacopa monnieri (Brahmi) on phencyclidine-induced disruptions in object recognition memory and cerebral calbindin, parvalbumin, and calretinin immunoreactivity in rats. Neuropsychiatr Dis Treat. (2019) 15:1103. doi: 10.2147/NDT.S193222

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Singh B, Pandey S, Rumman M, Kumar S, Kushwaha PP, Verma R, et al. Neuroprotective and neurorescue mode of action of Bacopa monnieri (L.) Wettst in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinson's disease: an in silico and in vivo study. Front Pharmacol. (2021) 12:169. doi: 10.3389/fphar.2021.616413

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Stough C, Lloyd J, Clarke J, Downey L, Hutchison C, Rodgers T, et al. The chronic effects of an extract of Bacopa monniera (Brahmi) on cognitive function in healthy human subjects. Psychopharmacology. (2001) 156:481–4. doi: 10.1007/s002130100815

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Stough C, Downey LA, Lloyd J, Silber B, Redman S, Hutchison C, et al. Examining the nootropic effects of a special extract of Bacopa monniera on human cognitive functioning: 90 day double-blind placebo-controlled randomized trial. Phytother Res. (2008) 22:1629–34. doi: 10.1002/ptr.2537

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Roodenrys S, Booth D, Bulzomi S, Phipps A, Micallef C, Smoker J. Chronic effects of Brahmi (Bacopa monnieri) on human memory. Neuropsychopharmacology. (2002) 27:279–81. doi: 10.1016/S0893-133X(01)00419-5

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Morgan A, Stevens J. Does Bacopa monnieri improve memory performance in older persons? Results of a randomized, placebo-controlled, double-blind trial. J Altern Complem Med. (2010) 16:753–9. doi: 10.1089/acm.2009.0342

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Peth-Nui T, Wattanathorn J, Muchimapura S, Tong-Un T, Piyavhatkul N, Rangseekajee P, et al. Effects of 12-week Bacopa monnieri consumption on attention, cognitive processing, working memory, and functions of both cholinergic and monoaminergic systems in healthy elderly volunteers. Evid Based Complem Altern Med. (2012) 2012:606424. doi: 10.1155/2012/606424

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Benson S, Downey LA, Stough C, Wetherell M, Zangara A, Scholey A. An acute, double-blind, placebo-controlled cross-over study of 320 mg and 640 mg doses of Bacopa monnieri (CDRI 08) on multitasking stress reactivity and mood. Phytother Res. (2014) 28:551–9. doi: 10.1002/ptr.5029

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Kean JD, Kaufman J, Lomas J, Goh A, White D, Simpson D, et al. A randomized controlled trial investigating the effects of a special extract of Bacopa monnieri (CDRI 08) on hyperactivity and inattention in male children and adolescents: BACHI study protocol (ANZCTRN12612000827831). Nutrients. (2015) 7:9931–45. doi: 10.3390/nu7125507

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Kumar N, Abichandani L, Thawani V, Gharpure K, Naidu M, Venkat Ramana G. Efficacy of standardized extract of Bacopa monnieri (Bacognize®) on cognitive functions of medical students: a six-week, randomized placebo-controlled trial. Evid Based Complem Altern Med. (2016) 2016:4103423. doi: 10.1155/2016/4103423

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Mishra M, Mishra AK, Mishra U. Brahmi (Bacopa monnieri Linn.) in the treatment of dementias–a pilot study. Fut Healthc J. (2019) 6:69. doi: 10.7861/futurehosp.6-1-s69

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Micheli L, Spitoni S, Di Cesare Mannelli L, Bilia AR, Ghelardini C, Pallanti S. Bacopa monnieri as augmentation therapy in the treatment of anhedonia, preclinical and clinical evaluation. Phytother Res. (2020) 34:2331–40. doi: 10.1002/ptr.6684

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Sanzini E, Badea M, Dos Santos A, Restani P, Sievers H. Quality control of plant food supplements. Food Funct. (2011) 2:740–6. doi: 10.1039/c1fo10112a

PubMed Abstract | CrossRef Full Text | Google Scholar

114. George P. Concerns regarding the safety and toxicity of medicinal plants-An overview. J Appl Pharm Sci. (2011) 1:40–4.

PubMed Abstract | Google Scholar

115. Balaji B, Kumar EP, Kumar A. Evaluation of standardized Bacopa monniera extract in sodium fluoride-induced behavioural, biochemical, and histopathological alterations in mice. Toxicol Ind Health. (2015) 31:18–30. doi: 10.1177/0748233712468018

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Singh D, Arya P, Koolwal N, Singh V, Saxena R, Sharma M, et al. Protective role of Bacopa monniera L. against hepatic oxidative stress in wistar albino rats. J Pharmacogn Phytochem. (2015) 4:233.

Google Scholar

117. Shahid M, Subhan F, Ullah I, Ali G, Alam J, Shah R. Beneficial effects of Bacopa monnieri extract on opioid induced toxicity. Heliyon. (2016) 2:e00068. doi: 10.1016/j.heliyon.2016.e00068

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Sumathi T, Devaraj SN. Effect of Bacopa monniera on liver and kidney toxicity in chronic use of opioids. Phytomedicine. (2009) 16:897–903. doi: 10.1016/j.phymed.2009.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Srivastav S, Fatima M, Mondal AC. Bacopa monnieri alleviates paraquat induced toxicity in Drosophila by inhibiting jnk mediated apoptosis through improved mitochondrial function and redox stabilization. Neurochem Int. (2018) 121:98–107. doi: 10.1016/j.neuint.2018.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Jyoti A, Sethi P, Sharma D. Bacopa monniera prevents from aluminium neurotoxicity in the cerebral cortex of rat brain. J Ethnopharmacol. (2007) 111:56–62. doi: 10.1016/j.jep.2006.10.037

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Velaga MK, Basuri CK, Robinson Taylor KS, Yallapragada PR, Rajanna S, Rajanna B. Ameliorative effects of Bacopa monniera on lead-induced oxidative stress in different regions of rat brain. Drug Chem Toxicol. (2014) 37:357–64. doi: 10.3109/01480545.2013.866137

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Ayyathan DM, Chandrasekaran R, Thiagarajan K. Neuroprotective effect of Brahmi, an ayurvedic drug against oxidative stress induced by methyl mercury toxicity in rat brain mitochondrial-enriched fractions. Nat Prod Res. (2015) 29:1046–51. doi: 10.1080/14786419.2014.968153

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Dwivedi S, Nagarajan R, Hanif K, Siddiqui HH, Nath C, Shukla R. (2013). Standardized extract of Bacopa monniera attenuates okadaic acid induced memory dysfunction in rats: effect on Nrf2 pathway. Evid Based Complem Altern Med. (2013) 2013:294501. doi: 10.1155/2013/294501

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Pham HTN, Phan SV, Tran HN, Phi XT, Le XT, Nguyen KM, et al. Bacopa monnieri (L.) ameliorates cognitive deficits caused in a trimethyltin-induced neurotoxicity model mice. Biolo Pharm Bull. (2019) 42:1384–93. doi: 10.1248/bpb.b19-00288

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Brimson JM, Prasanth MI, Plaingam W, Tencomnao T. Bacopa monnieri (L.) wettst. Extract protects against glutamate toxicity and increases the longevity of Caenorhabditis elegans. J Trad Complemen Med. (2020) 10:460–70. doi: 10.1016/j.jtcme.2019.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Sireeratawong S, Jaijoy K, Khonsung P, Lertprasertsuk N, Ingkaninan K. Acute and chronic toxicities of Bacopa monnieri extract in Sprague-Dawley rats. BMC Complement Altern Med. (2016) 16:249. doi: 10.1186/s12906-016-1236-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: pharmacological potential, tau aggregates, Bacopa monnieri, anti-cancer agents, neurodegenerative diseases

Citation: Fatima U, Roy S, Ahmad S, Ali S, Elkady WM, Khan I, Alsaffar RM, Adnan M, Islam A and Hassan MI (2022) Pharmacological attributes of Bacopa monnieri extract: Current updates and clinical manifestation. Front. Nutr. 9:972379. doi: 10.3389/fnut.2022.972379

Received: 18 June 2022; Accepted: 28 July 2022;
Published: 18 August 2022.

Edited by:

Carla Pereira, Centro de Investigação de Montanha (CIMO), Portugal

Reviewed by:

Con Stough, Swinburne University of Technology, Australia
Ufuk Okkay, Atatürk University, Turkey
Dharmendra Maurya, Bhabha Atomic Research Centre (BARC), India

Copyright © 2022 Fatima, Roy, Ahmad, Ali, Elkady, Khan, Alsaffar, Adnan, Islam and Hassan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Urooj Fatima, ufatima@jmi.ac.in; Md. Imtaiyaz Hassan, mihassan@jmi.ac.in

Download