Abstract
Neurodegenerative diseases like Alzheimer's disease and Parkinson's disease are severe disorders characterized by progressive neuron degeneration, leading to cognitive decline, motor dysfunction, and other neurological issues, significantly impairing daily life and the quality of life. Despite advancements in understanding these mechanisms, many aspects remain unclear, and current treatments primarily manage symptoms without halting disease progression. Multiple biological pathways are implicated in neurodegeneration, including oxidative stress, neuroinflammation, mitochondrial dysfunction, and aberrant protein folding. Given the multifactorial nature of neurodegenerative diseases, a neuroprotective approach targeting various mechanisms holds significant promise for prevention. Natural products derived from plants, animals, and fungi, known for their antioxidant and anti-inflammatory properties, show substantial potential in the prevention of neurodegeneration. Unlike synthetic compounds, bioactive compounds from these natural sources offer diverse targets due to their varied structures and biological activities. This review focuses on the potential of bioactive compounds from plants with sedative and mood-modulating effects in preventing and/or slowing down neurodegeneration.
Keywords: Phytobioactive compounds, neurodegeneration, oxidative stress, neuroinflammation, neuroprotection, Parkinson’s disease, Alzheimer’s disease
1. INTRODUCTION
Neurodegenerative diseases, such as Alzheimer’s Disease (AD) and Parkinson’s Disease (PD), are debilitating disorders of the nervous system characterized by progressive degeneration of neurons, resulting in gradual loss of cognitive function, motor control, and other neurological processes [1]. This condition significantly impairs daily functioning with devastating effects on the quality of life [2, 3]. After extensive years of research, substantial evidence points to multifactorial conditions as the roots of these disorders. These include 1) abnormal protein folding leading to defective protein degradation and aggregation, 2) oxidative stress triggering free radical formation, 3) impaired bioenergetics and mitochondrial dysfunction, and 4) exposure to metal toxicity and pesticides [4-6]. Despite extensive research aimed at unravelling the molecular mechanisms behind neurodegeneration, many aspects of these pathologies remain enigmatic. The current interventions for neurodegenerative diseases primarily focus on symptom management, with real disease-modifying treatments still under exploration.
AD is a progressive neurodegenerative disorder that primarily affects brain function, leading to memory loss and cognitive decline [7]. It is characterized by the accumulation of abnormal protein deposits in the brain, including beta-amyloid plaques (Aβ) and neurofibrillary tangles composed of hyperphosphorylated Tau protein (pTau) [8]. These pathological alterations result in neuron degeneration and disruption of synaptic transmission, particularly in the regions of the brain responsible for memory and cognitive functions [9]. As the disease progresses, individuals may experience difficulties with language, problem-solving, and performing familiar tasks.
PD is the most common age-related motoric neurodegenerative disease characterized by the gradual deterioration of dopaminergic neurons located in the substantia nigra pars compacta (SNpc) and the abnormal aggregation and accumulation of α-synuclein (α-syn) in the form of Lewy bodies, which accumulate within neurons disrupting normal cellular function and contributing to the progression of PD [10]. This neuronal loss in the SNpc results in the depletion of dopamine, which is a neurotransmitter critical for regulating movement [3, 11]. Consequently, the clinical features traditionally associated with PD are motor symptoms, including tremors, rigidity, bradykinesia, and postural instability. However, PD is now recognized to be a complex disorder involving a wide range of non-motor manifestations that contribute to disability.
Given the multifactorial nature of pathological mechanisms associated with neurodegeneration, adopting a neuroprotection approach that targets various mechanisms could be a promising strategy for the prevention of neurodegenerative diseases. Over the past few years, there has been a surge of interest in natural products derived from plants, animals, and fungi, along with their bioactive compounds, due to their inherent antioxidant and anti-inflammatory properties, which hold significant potential in preventing and/or delaying neurodegeneration. Interestingly, bioactive compounds offer a wider range of targets compared to chemical compounds due to their varied structures and biological activities [12].
In this review, we focused on the potential benefits of bioactive compounds extracted from plants possessing sedative and mood-modulating effects in the context of neurodegenerative disorders. These compounds refer to chemical substances found in certain plant species that have physiological effects on the body, particularly in promoting relaxation, reducing anxiety, and improving mood. Although these plants are renowned for their calming and relaxing effects, there is growing attention to their potential neuroprotective properties against neurodegeneration. These compounds, encompassing flavonoids, terpenoids, alkaloids, and phenolic compounds, are significant for their capacity to reduce oxidative stress and inflammation, both of which are implicated in the onset and progression of neurodegenerative disorders, such as AD and PD (Fig. 1) [13].
Fig. (1).

Progression of neurodegenerative disorders.
2. MECHANISMS AND POTENTIAL TARGETS OF NEURODEGENERATION
Neurodegenerative disorders encompass a complex interplay of various pathological mechanisms, each contributing to the progressive decline in neuronal function and viability. Among these mechanisms, oxidative stress arises from an intricate imbalance between the production of free radicals and the capacity of antioxidant defenses within cells [14]. Free radicals, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS), are highly reactive molecules generated as natural byproducts of cellular metabolism, especially within mitochondria, as well as in response to environmental stressors [15]. Under normal circumstances, cells possess an efficient repertoire of antioxidant enzymes, including superoxide dismutase, catalase, and glutathione peroxidase, along with non-enzymatic antioxidants like vitamins C and E, which work synergistically to neutralize these harmful radicals and maintain redox homeostasis [16]. However, in the context of neurodegenerative diseases, this delicate balance is disrupted, resulting in an overwhelming accumulation of free radicals that surpass the scavenging capacity of endogenous antioxidants [14, 17]. Consequently, these unchecked radicals indiscriminately target cellular macromolecules, including lipids, proteins, and nucleic acids, leading to oxidative damage and functional impairment [18]. Moreover, oxidative stress serves as a powerful catalyst, amplifying neurodegenerative processes by triggering a cascade of events that worsens cellular dysfunction and promotes cell death [14]. For instance, oxidative modification of proteins can promote misfolding and aggregation, contributing to the formation of toxic protein aggregates that characterize both AD and PD pathogenesis [17, 19-22]. Additionally, oxidative stress can further exacerbate mitochondrial dysfunction, leading to impaired energy metabolism and increased ROS production, thus perpetuating a vicious cycle of cellular damage and dysfunction [23-25]. Dysfunction in mitochondrial activity, characterized by compromised energy production and increased ROS generation, significantly contributes to neurodegenerative processes, including metabolism dysfunction [17, 26]. However, the consequences of mitochondrial dysfunction extend beyond impaired energy metabolism and oxidative stress. Indeed, since mitochondria are crucial regulators of calcium (Ca2+) homeostasis, their dysfunction can disrupt intracellular Ca2+ signaling, leading to neuronal excitotoxicity and cell death [17, 27]. Moreover, dysfunctional mitochondria are unable to adequately regulate apoptotic pathways, resulting in increased susceptibility to programmed cell death [28]. Another critical factor involved in the pathogenesis of neurodegenerative disorders is neuroinflammation. It is characterized by the activation of various immune cells within the Central Nervous System (CNS), including microglia and astrocytes, in response to pathological stimuli [29]. This inflammatory response is a double-edged sword, as while it is initially intended to protect the brain from harmful insults, prolonged or dysregulated activation can lead to detrimental consequences [30]. Indeed, prolonged activation of microglia and astrocytes triggers the release of pro-inflammatory cytokines, chemokines, and ROS, which not only exacerbate neuronal damage but also disrupt the delicate balance of neurotransmitters and neurotrophic factors essential for physiological neural function [31]. This disruption in neural signaling pathways further fuels the progression of neurodegeneration, creating a vicious cycle of neuronal injury and inflammation [32, 33]. Furthermore, neuroinflammation is intricately linked with oxidative stress and protein aggregation, contributing to amplifying the detrimental effects of neuroinflammation on neuronal health and accelerating disease progression [34].
Considering the multifaceted nature of neurodegeneration, pursuing a strategy that targets multiple mechanisms of action holds promise for both preventing and treating neurodegenerative diseases. It is noteworthy that bioactive compounds act directly on these pathological mechanisms, offering neuroprotective effects by attenuating oxidative stress, restoring mitochondrial function, and dampening inflammatory responses.
3. PLANT-DERIVED BIOACTIVE COMPOUNDS
Phytobioactive compounds are plant-derived substances that offer protection against various diseases, yet they are not essential components of our diet. These substances, characterized by unique structural and functional properties, are commonly known as phytochemicals or secondary metabolites of plants [35]. These compounds include flavonoids, terpenoids, alkaloids, and phenolic compounds.
Polyphenols are a diverse group of phytochemicals found in plant-based foods, such as fruits, vegetables, nuts, seeds, and beverages like tea and red wine. They are characterized by their multiple phenol rings and can be categorized into subclasses, such as flavonoids, phenolic acids, and stilbenes [36]. One of the key properties of polyphenols is antioxidant activity, which involves scavenging free radicals and enhancing the expression of antioxidant enzymes, such as γ-glutamylcysteine synthetase (γGCS), glutathione peroxidase (GPX), superoxide dismutase (SOD), and glutathione reductase (GSH). This activity helps to reduce the risk of various chronic diseases, such as neurodegenerative disorders [37]. Moreover, polyphenols exhibit a range of other beneficial effects beyond their antioxidant properties, including anti-inflammatory activity [38]. Different polyphenolic compounds may vary in their antioxidant and anti-inflammatory capacity, depending on factors, such as their chemical structure, concentration, and bioavailability [12].
Flavonoids, a subgroup of polyphenolic compounds abundantly found in fruits (apples, berries, cherries [39]), vegetables (onions [39]), and medical plants (chamomile [40]), have garnered considerable attention for their potential neuroprotective properties [41]. Flavonoids exhibit anti-inflammatory properties by inhibiting the production and release of pro-inflammatory mediators, such as cytokines and chemokines, and suppressing the activation of microglia and astrocytes, thereby slowing neurodegeneration. Flavonoids also modulate key molecular targets involved in neurodegenerative processes, including the inhibition of Aβ aggregation in AD and the modulation of α-syn accumulation in PD [42, 43]. These compounds also promote neurogenesis and enhance synaptic plasticity, which is essential for learning and memory [44, 45]. Their antioxidant and anti-inflammatory properties help protect neurons from oxidative damage and neuroinflammation while also supporting neuronal survival by influencing signaling pathways related to apoptosis, neurogenesis, and synaptic function [46]. Beyond their antioxidant and anti-inflammatory actions, phenolic compounds are also involved in the inhibition of toxic protein aggregation in both AD and PD [47] and in the promotion of neuronal survival by modulating signaling pathways involved in apoptosis, neurogenesis, and synaptic plasticity [48]. Research highlights the neuroprotective actions of flavonoids, such as nobiletin, fisetin, and cocoa, in neurodegenerative disorders [40]. Nobiletin, derived from citrus peels, shows promise in treating AD and PD by reducing Aβ plaque and pTau accumulation, alleviating oxidative stress, and improving both motor and cognitive functions [49]. Fisetin, found in strawberries, exerts a neuroprotective role in PD by boosting antioxidant defenses, supporting cognitive function, and increasing dopamine levels [50]. Cocoa, rich in flavonoids like flavan-3-ols, helps mitigate Aβ-induced neurotoxicity, preserves cell viability, and supports neuronal network integrity. Its antioxidant and anti-inflammatory properties make it a potential candidate for attenuating neurodegeneration [51]. Additionally, cocoa offers cardiovascular benefits by reducing arterial stiffness, lowering triglycerides, enhancing nitric oxide synthase activity [52], and reducing oxidative stress and platelet reactivity [53].
Terpenoids, also known as terpenes, are aromatic compounds found in plants (salvia) [54], which possess diverse pharmacological properties. They have demonstrated promise in enhancing cognitive function, reducing neuroinflammation, and protecting against neuronal damage in neurodegenerative diseases. In AD, terpenoids may help preserve neuronal function and prevent the formation of Aβ plaques and tau tangles [55]. Similarly, in PD, they could protect dopaminergic neurons in the substantia nigra, potentially slowing the progression of the disease [56]. Another property of terpenoids is the modulation of neurotransmitter levels and activity. For instance, they may enhance cholinergic function in AD by inhibiting acetylcholine breakdown [57] and improve motor symptoms in PD by affecting dopamine levels or receptor sensitivity [58]. Notably, terpenoids can cross the blood-brain barrier, allowing them to exert their neuroprotective effects directly within the central nervous system, making them valuable for treating conditions like AD and PD [59].
An important terpenoid-based treatment is derived from Astragalus membranaceus (A. membranaceus), a plant well-known for its neuroprotective properties. Studies indicate that A. membranaceus significantly enhances recovery from hemorrhagic stroke by reducing brain edema and improving patient outcomes through its antioxidative and anti-inflammatory effects [60]. Additionally, Astragaloside IV (AS-IV), a compound from A. membranaceus, has shown neuroprotective benefits across several neurological disorder models. AS-IV reduces blood-brain barrier permeability, lymphocyte infiltration, neuroinflammation, apoptosis, and oxidative stress. In AD models, it inhibits Aβ production and plaque formation, while in PD, it promotes neuron survival by enhancing antioxidant defenses [61].
Alkaloids are nitrogen-containing compounds found in many plants, and most of them have definite pharmacological properties [35]. Their antioxidative properties make them promising candidates for treating neurodegenerative diseases by addressing oxidative stress. Studies have shown that alkaloids demonstrate anti-inflammatory properties by inhibiting various pro-inflammatory protein complexes [62], including NF-κB, ERK1/2, Akt, and STAT1, as well as inflammatory mediators like prostaglandin E2, nitric oxide, cytokines, and chemokines [63, 64]. Interestingly, alkaloids can improve the pathophysiology of neurodegenerative disorders by functioning as inhibitors of monoamine oxidase (MAO), acetylcholinesterase, and butyrylcholinesterase and by acting as N-methyl-D-aspartate (NMDA) antagonists, as well as agonists of muscarinic and adenosine receptors [65].
The diverse mechanisms of action of these bioactive compounds make them appealing candidates for preventing neurodegenerative diseases.
In the following paragraph, we will discuss how natural products and their bioactive compounds show promise in alleviating neurodegenerative processes (Fig. 2).
Fig. (2).
The diverse mechanisms of action of these bioactive compounds.
4. NEUROPROTECTIVE EFFECTS OF NATURAL PRODUCTS AND THEIR BIOACTIVE COMPOUNDS IN AD AND PD
4.1. Griffonia simplicifolia
G. simplicifolia is a climbing leguminous plant belonging to the family of Fabaceae, native to West Africa, especially in Ghana, Liberia, Togo, and the Ivory Coast. Phytochemical analyses demonstrated that this plant contains several active compounds as well as non-proteinogenic amino acids, such as 5-hydroxytryptophan, a known precursor of serotonin [66]. G. simplicifolia may be considered an effective natural alternative to selective serotonin reuptake inhibitors and synthetic antidepressants for the treatment of mood disorders, anxiety, and depression [67], and it is also used as a secondary treatment for sleep disorders and appetite [68, 69]. Furthermore, G. simplicifolia exerts several biological actions, including antioxidant, anti-inflammatory, antibacterial, and anticancer activities, due to tannins, flavonols, and carbolide alkaloids [70]. Gueyraud et al. explored the antioxidant potential of the aqueous leaf extract of G. simplicifolia [71]. They first demonstrated that the viability of neural cells was not compromised with an extract concentration of up to 800 μg/mL, while the concentration of 20 μg/mL was able to protect neurons and astrocytes from oxidative stress induced by hydrogen peroxide [71]. Regarding the seed extracts, a metabolomic study on different used solvents highlighted acetone and ethanol as optimal for the extraction of flavonoids and flavan-3-ols, while microwave-assisted extraction displayed enhanced effectiveness in extracting N-containing compounds, including 5-hydroxytryptophan. Furthermore, this analysis demonstrated that the hydroalcoholic extracts exhibited the highest radical scavenging and metal-reducing antioxidant power [72].
4.2. Sambucus nigra
S. nigra is an herbaceous species of the Caprifoliaceae family, and it spontaneously grows in Europe, West Asia, and North America. Although its leaves and stems are toxic due to their content of cyanogenic glycosides (sambunigrin and prunasin) and m-hydroxysubstituted glycosides (zierin and holocalin), which, after hydrolysis, can release cyanide [73], the elderflower and fruits are used as herbal medicine and for food purposes [74-76]. Quercetin, rutin, and kaempferol, potent flavonoids, and chlorogenic, caffeic, and protocatechuic acids, known as hydroxycinnamic acids, give elderflower its antibacterial [77], immunomodulatory [78], diuretic and anti-diabetic properties [79]. The antioxidant and protective potential of elderflower were confirmed in the human neuroblastoma SH-SY5Y cell line [80]. The S. nigra flower extract also exhibited the ability to regulate mTORC1 signaling activity, and therefore, it positively regulated the pathogenic effects of molecular mechanisms involved in neurodegenerative diseases [80].
4.3. Grewia tiliaefolia
G. tiliaefolia is a subtropical tree specifically found in the Himalayan tract from Jammu to Assan, the central Vindya range, and the Western and Eastern Ghats of India [81]. In Indian traditional medicine, the plant is used as an astringent, expectorant, antipruritic, and aphrodisiac [82]. The Grewia genus consists of 150 species and is a member of the family Tiliaceae found in tropical and subtropical areas; about 40 of these species are found in India [83]. The methanol extract of G. tiliaefolia has shown significant free radical scavenging properties and an electron-donating ability and, therefore, can be considered an effective antioxidant [82]. Since different antioxidants have demonstrated protective effects against the neurodegenerative spiral induced by Alzheimer's redox imbalance [25, 84], methanol extracts of G. tiliaefolia may be protective as well. Leaf extracts also showed the ability to inhibit cholinesterase enzymes. Notably, polar extracts (methanol and water) showed higher inhibition compared to non-polar solvent extracts. Various compounds of methanol extract, such as vitexin, vitexin-4-O-glucoside, 3-O-methyl ellagic acid, isovitexin, and nitidanin, demonstrated the ability to interact with various docking sites present on the AchE enzyme. Of note, vitexin showed significant and dose-dependent inhibition of both AchE and BchE [82, 83]. Moreover, a methanol extract of G. tiliaefolia managed to protect Neuro2a cells from the cytotoxic effects of Aβ25-35 while also restoring cell viability [82]. Malar et al. reported that Aβ25-35 leads to an increase in fluorescence intensity, indicating the formation of oligomers and aggregates. Treatment with methanol extract of G. tiliaefolia managed to reduce the intensity of fluorescence similarly to galantamine (50 µM). Therefore, the methanol extract managed to reduce Aβ25-35 aggregation and fibril formation [82].
4.4. Passiflora incarnata
P. incarnata, commonly known as the passionflower, belongs to the Passifloraceae family, and it is a flowering plant renowned for its medicinal properties. P. incarnata is native to the southeastern United States, particularly in regions like Florida and Texas, as well as parts of Central and South America. The main constituents of the leaves of P. incarnata are flavonoids, maltol, and indole alkaloids [85]. Historically, the pharmacological investigations of P. incarnata primarily focused on its sedative, anxiolytic, and anticonvulsant impacts on the CNS, alongside its potential to aid in the treatment of addiction [86-89]. Later on, further research highlighted the neuroprotective properties of P. incarnata, shedding light on its potential to mitigate the loss of neurons in PD and AD, thereby preserving cognitive function and motor abilities in affected individuals [90]. It has been reported that the most promising neuroactive substances are flavonoids, such as chrysin, vitexin, isovitexin, orientin, isoorientin, apigenin, and kaempferol, as well as indole alkaloids [91]. In a study conducted by Kim et al., vitexin was identified as the active compound in the extract of P. incarnata, showing a diverse array of biological functions, including antioxidant, anti-inflammatory, and anti-AD properties [92]. Notably, the study reported substantial enhancements in neurocognitive function and reductions in pTau/Tau levels following treatment with P. incarnata extract [92]. P. incarnata also demonstrated antiparkinsonian and memory-enhancing effects, probably due to its high phenolic and flavonoid content [90]. Specifically, the butanol extract of P. incarnata exhibited significant reductions in haloperidol-induced catalepsy, tacrine-induced jaw movements, and the overproduction of free radicals [90]. A recent study reported that the extract of P. incarnata significantly impacts dopaminergic neurotransmission and dopamine metabolism in the CNS [93]. In this regard, an increased level of dopamine and its metabolites was observed in the spinal cord following treatment with an extract of P. incarnata, suggesting its potentially beneficial effect on PD, which is characterized by a deficit in dopaminergic neurotransmission [93].
4.5. Crataegus oxyacantha
C. oxyacantha, also known as Hawthorn, is a shrub of the Rosaceae family that can be found in the hilly areas of Himachal Pradesh and Kashmir [94]. This plant is a rich source of triterpenic acids, ursolic acid, oleanolic acid, polyphenols, flavonoids, glycosides, and other important organic molecules [95]. Due to this composition, C. oxyacantha is used to treat many cardiovascular diseases [96, 97], as well as hypertension, arthritis, and hyperlipidaemia [98]. Another important role of C. oxyacantha is in the prevention of neurodegenerative disorders, such as AD and dementia, as well as attention deficit disorder and anxiety [99]. Indeed, extracts of this fruit can be used as natural antimicrobial and antioxidant preparations [100]. Different studies have demonstrated the ameliorative role of C. oxyacantha in cognitive impairment and brain oxidative damage [101] and its possible use in AChE and BChE inhibitions [99]. C. oxyacantha also demonstrates an ability to enhance learning and memory. This is observed through a reduction in the transfer latency time in rats with scopolamine-induced amnesia on an elevated maze. Furthermore, it helps restore the balance of oxidative stress markers in the brain that are disturbed due to the administration of scopolamine. Additionally, it inhibits serotonergic transmission while promoting dopaminergic transmission [102]. In conclusion, C. oxyacantha is a rich source of neurologically potent organic molecules, which play a vital role in the attenuation of neurodegeneration and its related nervous system and neuroprotective effect.
4.6. Valeriana officinalis
V. officinalis, a perennial herb belonging to the Caprifoliaceae family, is widely distributed across America, Europe, and Asia [103]. Among approximately 200 identified species, V. officinalis is particularly notable for its medicinal applications [104]. The bioactive constituents of V. officinalis, including valerenic acid, valerenal, and various antioxidants, contribute to its efficacy as a natural remedy for anxiety and sleep disorders [105]. These compounds interact with neurotransmitter receptors in the brain, such as gamma-aminobutyric acid (GABA) receptors, promoting relaxation and reducing nervous tension. In addition to its traditional use as a sedative, recent research has explored its potential in addressing neurodegenerative diseases like AD and PD [106]. Studies indicate that valerian extracts may possess neuroprotective effects by mitigating oxidative stress and inflammation, both of which are implicated in the progression of these pathological conditions. V. officinalis boasts a rich chemical composition, comprising over 150-200 chemical constituents [107]. Among these, flavonoids renowned for their effects on the CNS, as well as lignans with antioxidative and vasorelaxant properties [108]. De Oliveira and colleagues demonstrated that, in an in vitro model of PD, the aqueous extract of V. officinalis provides cytoprotection against rotenone-induced apoptosis in SH-SY5Y cells, suggesting a potential neuroprotective effect. This outcome is attributed to the antioxidant properties associated with lignans found in V. officinalis. Indeed, the generation of ROS and subsequent oxidative stress resulting from the inhibition of mitochondrial complex I induced by rotenone seem to play a significant role in the PD model based on rotenone toxicity [109]. Another study conducted by Malva and colleagues found that V. officinalis has been associated with a GABAergic mechanism [110]. This study aimed to assess its neuroprotective potential against Aβ 25-35 toxicity in primary rat hippocampal neurons. It was found that V. officinalis extract prevented neuronal injury induced by Aβ25-35, probably by inhibiting excess Ca2+ influx and membrane peroxidation. These findings highlighted the neuroprotective properties of V. officinalis extract against Aß toxicity, offering potential applications in aging and neurodegenerative disorders [110]. Marawne and colleagues investigated the effects of V. officinalis on glial cells under conditions mimicking neurodegenerative inflammation commonly observed in diseases, such as AD and PD [111]. In particular, they observed that the alcoholic extract of V. officinalis reduced microglial activation, NO production, and gene expression of Tumor Necrosis Factor α (TNF-α) and induced nitric oxide synthase (iNOS). Notably, lower concentrations of the extract mitigated inflammation, while higher doses promoted cell survival. Moreover, the secondary metabolites of V. officinalis, including valeric acid and valepotriates, were associated with relaxing, sleep-inducing, and neuroprotective properties. Specifically, valerenic acid exhibited potential in mitigating neurodegeneration in a mouse model of PD, potentially preserving dopaminergic neurons and restoring motor function. Additionally, valproic acid showed anti-inflammatory effects, reducing pain responses in mice and safeguarding against oxidative stress [111].
4.7. Rhodiola rosea
R. rosea, commonly known as the golden or arctic root, is a perennial flowering plant native to the Arctic regions of Europe, Asia, and North America. This plant belongs to the Crassulaceae plant family, specifically to the subfamily Sedoideae and the genus Rhodiola [112]. This botanical plant has been traditionally used to invigorate the nervous system, alleviate depression, improve work performance, and prevent altitude sickness. Among the various species within the Rhodiola genus, R. rosea has garnered considerable attention for its phytochemical composition and toxicological properties. The root of R. rosea contains approximately 28 compounds, with salidroside (rhodioloside), rosavins, and p-tyrosol, which are believed to exhibit the most significant beneficial effects. Research suggests that ingestion of R. rosea can enhance cognitive function, reduce mental fatigue, mitigate free radicals, boost endurance performance, and improve learning and memory [112]. Furthermore, there is a growing interest in the potential of R. rosea to mitigate neurodegenerative conditions like AD [113]. Its anti-inflammatory properties and neuroprotective abilities present promising avenues for exploring treatments for AD. Lee and colleagues investigated the anti-inflammatory and neuroprotective properties of constituents from R. rosea in both microglial and neuronal cells [112]. Microglia, known for their pivotal role in inflammatory reactions, are implicated in neurodegenerative pathologies like AD and PD, where their activation contributes to the production of harmful ROS and proinflammatory cytokines. Studies reported that R. rosea effectively inhibits the production of NO and the expression of iNOS in microglial cells stimulated with lipopolysaccharides (LPS), a known inflammatory stimulus. Additionally, R. rosea treatment reduced the levels of proinflammatory cytokines TNF-α, IL-1β, and IL-6 induced by LPS in microglial cells. Another study observed that oral administration of R. rosea extract decreased iNOS and proinflammatory cytokine responses in both the kidney and prefrontal cortex in mice, indicating its potential role in suppressing inflammation in the CNS. Furthermore, R. rosea demonstrated protective effects against L-glutamate-induced neurotoxicity in cortical neurons by inhibiting JNK and p38MAPK activation, underlining its neuroprotective potential against neurodegenerative diseases [112]. The pretreatment effects of R. rosea extract on cognitive dysfunction, oxidative stress, and hippocampal neuron injury in a rat model of AD were investigated by Qu et al. [114]. This study focused on the potential of R. rosea to exert a protective effect against AD-related deficits, particularly by reducing oxidative stress and improving cognitive function and neuronal damage. These findings suggest that R. rosea could potentially mitigate AD-associated cognitive deficits through its antioxidative properties.
4.8. Humulus lupulus
H. lupulus, commonly known as hops, is a perennial climbing plant known for its role in brewing beer, originating from Europe, Asia, and North America. With its distinctive cones, this member of the Cannabaceae family imparts bitterness, flavour, and aroma to the beverage [115]. Beyond brewing, hops have been used for centuries in traditional medicine for their sedative properties. Recently, H. lupulus has gained attention for its potential neuroprotective effects against neurodegenerative diseases [116]. Several studies have suggested that certain compounds found in hops, such as xanthohumol and its derivatives, possess antioxidant and anti-inflammatory properties that could help mitigate the progression of these debilitating conditions [116]. Additionally, hop extracts have shown promise in modulating neurotransmitter levels and improving cognitive function in experimental models of neurodegeneration [117]. A recent study conducted by Palmioli et al. explored the impact of hop extracts on the aggregation and toxicity of synthetic Aβ1-42 peptides [118]. Their investigation revealed the antioxidant capacity of hop extracts and their ability to promote autophagy, facilitating the clearance of amyloid aggregates. This study investigated the antioxidant characteristics of hop extracts, by testing them on SH-SY5Y cells to evaluate their antioxidant effects under oxidative stress conditions. Notably, hop extracts were found to not only inhibit the aggregation of Aβ1-42 but also to shield against Aβ-induced toxicity in vitro. Additionally, the polyphenols found in hop extracts emerged as potent anti-amyloidogenic agents, showing significant antioxidant properties [118]. Another study reported that the natural product extracted from H. lupulus inhibits monoacylglycerol lipase activity, reduces neurodegeneration, prevents neuroinflammation, and improves cognitive functions in AD animals [119].
4.9. Chamomilla matricaria
C. matricaria is a small flowering herbaceous plant belonging to the Asteraceae family [120]. Native to Europe, Asia, and North Africa, C. matricaria is characterized by its delicate, daisy-like flowers with white petals and yellow centers. This aromatic plant has a long history of use in various cultures for its medicinal, culinary, and cosmetic purposes [120]. C. matricaria is recognized for its calming and soothing properties, making it a popular ingredient in herbal teas, skincare products, and natural remedies. In addition to its traditional uses, C. matricaria has garnered attention for its potential beneficial effects in neurodegenerative diseases. Studies suggest that C. matricaria possesses antioxidant and anti-inflammatory properties, which may have neuroprotective effects [121]. These characteristics are due to the rich chemical composition of C. matricaria, such as α-bisabolol, chamazulene, bisabolol oxide A and B, matricin, and flavonoids like apigenin and luteolin [122]. In particular, α-bisabolol is renowned for its anti-inflammatory, antimicrobial, and skin-soothing properties, chamazulene possesses anti-inflammatory and antioxidant effects, and flavonoids contribute to the antioxidant properties of C. matricaria, and it may help alleviate inflammation and promote relaxation [123]. In a recent study conducted by Alahmady et al., the neuroprotective properties of C. matricaria were investigated in the context of AD [124]. It was found that, in rats exposed to aluminum chloride to induce cognitive impairment, an essential oil extracted from C. matricaria exhibited neuroprotective effects on the cholinergic system by increasing acetylcholine (ACh) levels and reducing inflammatory cytokines and apoptosis markers [124]. Moreover, C. matricaria extract exhibited antioxidant properties by protecting the rat brain from the increased oxidative stress caused by scopolamine administration, which is known to impair long-term potentiation [125]. These findings suggest that chamomile may offer a promising, cost-effective, and safe alternative to conventional AD therapy.
4.10. Melissa officinalis
M. officinalis, commonly known as lemon balm, is a perennial herbaceous plant belonging to the Lamiaceae family [126]. With a rich history dating back centuries, lemon balm has been revered for its medicinal properties and aromatic qualities. Native to the Mediterranean region and parts of Asia, this herb is now cultivated worldwide [126]. Rich in bioactive compounds like rosmarinic acid, flavonoids, and essential oils, lemon balm has been traditionally used to alleviate stress, aid digestion, and promote skin health [127-129]. Ongoing research explores its potential as an anti-inflammatory, antioxidant, and neuroprotective agent [129]. A study conducted by Ozarowski et al. examined the effects of prolonged administration of a standardized extract of M. officinalis on memory impairment induced by scopolamine in rats [130]. The study found that M. officinalis extract, containing rosmarinic acid, enhanced long-term memory in rats without affecting memory impairment [130]. Interestingly, the effects of the M. officinalis extract were studied in patients with mild to moderate AD [131]. It was observed that AD-affected patients showed significant cognitive benefits after 16 weeks of treatment with M. officinalis extract. Additionally, M. officinalis essential oil was found to positively affect agitation levels and the quality of life in patients with severe dementia [131]. M. officinalis has also been studied for the treatment of PD. In this regard, Martins and colleagues investigated the potential of M. officinalis extract in reducing manganese (Mn)-induced oxidative stress in mice, which can lead to neurodegenerative disorders resembling PD [132]. Of note, treatment with M. officinalis extracts significantly enhanced antioxidant enzyme activities and attenuated oxidative damage in the brain by replenishing the functions of superoxide dismutase and catalase enzymes, suggesting its potential antioxidative properties in mitigating Mn-induced oxidative stress [132].
4.11. Curcuma longa
Curcumin is the major component of C. longa, belonging to the Zingiberaceae family, which is a perennial herb widely cultivated in tropical regions of Asia [133]. Traditionally, curcumin has been used over the centuries for cooking as well as a phytotherapeutic agent [134]. Curcumin helps to maintain the homeostasis of the inflammatory system, improves the clearance of toxic aggregates from the brain, and induces antioxidant response elements [135-137]. However, the activity of curcumin is limited due to its low water solubility, fast biological metabolism, and limited blood-brain barrier (BBB) permeabilization [138]. To overcome this problem, various curcumin delivery systems have been developed based on nanoparticles. In PD drosophila and mice models, curcumin nanopreparation showed some anti-parkinsonism effects [139]. Specifically, in vitro results demonstrated that both α-syn oligomerization and fibrillation were significantly inhibited by curcumin, and that toxicity, oxidative stress, and cell death caused by rotenone were reduced [139]. Furthermore, studies reported that curcumin ameliorates behavioral damage in various AD models [140]. Curcumin is able to reverse neurotoxic damage induced by Aβ by preventing its aggregation and tau protein hyperphosphorylation, increasing mitochondrial ATP, and enhancing cytochrome oxidase activity [141].
4.12. Mucuna pruriens
M. pruriens, commonly known as velvet beans, belongs to the Fabaceae family and is native to India and the southern part of China [142]. It possesses valuable medicinal properties, including neuroprotective and antioxidant actions [143]. The major phenolic constituent of M. pruriens beans is the dopamine precursor, levodopa [144], which may help in ameliorating different types of free radical-mediated diseases like diabetes, arthritis, aging, and various other neurological disorders, such as PD [142]. Interestingly, the seeds of M. pruriens are rich in NADH and coenzyme Q10, which have been observed to exert beneficial effects in PD [145]. Animals treated with M. pruriens demonstrated significant improvement in behavioral tests along with reduced nerve damage and neuronal loss [146]. These findings suggest that M. pruriens could serve as an effective complementary treatment for PD, potentially enhancing the quality of life of the individuals affected by this disease [146]. Furthermore, treatment with the methanolic extract of M. pruriens significantly reversed memory deficits caused by ischemia [147]. This finding underscores its potential as a therapeutic option for severe brain injuries and neurodegenerative diseases.
5. METHODS
To select papers for this narrative review, we performed searches on PubMed and Google Scholar. The date of the most recent search was 21st May, 2024. The following terms were used in the searches: Alzheimer, Alzheimer’s disease, Parkinson, Parkinson’s disease, oxidative stress, mitochondrial dysfunction, protein folding, sedative plants, mood-modulating plants, neuroprotection, neurodegeneration, phytobioactive compounds, Griffonia simplicifolia, Sambucus nigra, Grewia tiliaefolia, Passiflora incarnata, Crataegus oxyacantha, Valeriana officinalis, Rhodiola rosea, Humulus lupulus, Chamomilla matricaria, Melissa officinalis, Curcuma longa, and Mucuna pruriens.
6. LIMITATIONS AND CHALLENGES ASSOCIATED WITH THE USE OF NATURAL PRODUCTS AND THEIR BIOACTIVE COMPOUNDS IN AD AND PD
As detailed in the present work, there is a large number of preclinical studies investigating and corroborating the beneficial effects of natural products and their bioactive compounds on AD and PD. However, we also have to take into account the various limitations of a phytotherapeutic approach.
Indeed, plant extracts usually contain different active biochemical compounds, each capable of exerting activity on various molecular targets. Furthermore, nutraceuticals derived from plants usually have a low and highly variable bioavailability, while herbal drug composition is usually inconsistent [148]. Inter-individual variability is another challenge to the effective use of plant extracts in clinical practice; age, sex, microbiome, genetic differences, and diet can influence the organism's response to such nutraceuticals [149]. Therefore, each promising compound should be isolated and purified to a high degree or artificially synthetized. Its individual efficacy must then be evaluated first on cellular models, then on animal models, and, ultimately, in human trials. Lastly, plant extracts and natural compounds can negatively interact with a vast array of drugs [150], potentially reducing their absorption (and therefore their bioavailability), their elimination or activation (by inhibiting their secretion or their metabolism), and can even lead to increased production of toxic metabolites (Table 1).
Table 1.
List of the main bioactive compounds from sedative and mood-modulating plants and their potential beneficial effects.
| Plants | Bioactive Compounds | Potential Beneficial Effects |
Results Obtained
(Theoretical or Experimental) |
References |
|---|---|---|---|---|
|
Griffonia
simplicifolia |
Saponins (secondary metabolites) 5-hydroxytryptophan |
Antioxidant | Experimental | [67, 68] |
|
Sambucus
nigra |
Flavonoids Phenolic Compounds (Hydroxycinnamic acids) |
Antioxidant | Experimental | [76] |
|
Grewia
tiliaefolia |
Flavonoids (Vitexin) | Antioxidant Anti-aggregating properties of abnormal proteins |
Experimental | [78] |
|
Passiflora
incarnata |
Flavonoids (Vitexin) | Antioxidant Anti-inflammatory Anti-aggregating properties of abnormal proteins |
Experimental | [86, 88, 89] |
|
Crataegus
oxyacantha |
Triterpenoid (Oleanolic and Ursolic Acid) Phenolic compounds, Flavonoids |
Antioxidant | Theoretical/ Experimental |
[95-98] |
|
Valeriana
officinalis |
Valerenic Acid, Valerenal and Valepotriates (secondary metabolites) Flavonoids | Antioxidant Anti-inflammatory Anti-aggregating properties of abnormal proteins Restoration of Mitochondrial Function |
Experimental | [105-107] |
|
Rhodiola
rosea |
Glycoside of Tyrosol (Rhodioloside) Phenolic Compounds (Rosavins) Phenolic Compounds (P-tyrosol) |
Antioxidant Anti-inflammatory |
Experimental/ Theoretical |
[108-110] |
|
Humulus
lupulus |
Prenylated flavonoid (Xanthohumol; 8-Prenylnaringenin) Phenolic Compounds (Flavan-3-ol Glycosides) Polyphenols (Procyanidins) |
Antioxidant Anti-inflammatory Anti-aggregating properties of abnormal proteins |
Theoretical/ Experimental |
[114, 115] |
| Chamomilla matricaria | Sesquiterpene alcohol (α-Bisabolol) Sequiterpene (Camazulene) Sequiterpene oxide (Bisabolol Oxide A) Hydroxycinnamic acid (Chlorogenic Acid) Flavonoids (Apigenin-7-glucoside, Rutin, cynaroside, Luteolin, apigenin and derivatives of Apigenin-7-glucoside) |
Antioxidant Anti-inflammatory |
Experimental | [120, 121] |
|
Melissa
officinalis |
Polyphenolic Compound Flavonoids | Antioxidant Anti-inflammatory |
Experimental | [126-128] |
|
Curcuma
longa |
Polyphenolic Compound (Curcumin) | Antioxidant Anti-inflammatory Anti-aggregating properties of abnormal proteins Restoration of Mitochondrial Function |
Theoretical/ Experimental |
[135-137] |
CONCLUSION
Neurodegenerative diseases, such as AD and PD, present profound challenges to an individual’s quality of life, primarily due to their progressive nature and the current lack of disease-modifying treatments. Despite extensive research efforts, the underlying molecular mechanisms are still not completely understood. However, a multifactorial approach targeting various pathways implicated in neurodegeneration offers promise. There is a growing interest in natural bioactive compounds derived from plants, animals, and fungi, known for their antioxidant and anti-inflammatory properties. In this review, we explored the neuroprotective potential of bioactive compounds extracted from plants possessing sedative and mood-modulating effects in mitigating oxidative stress, inflammation, mitochondrial dysfunction, and aberrant protein folding, which are critical factors involved in the pathogenesis of both AD and PD. This emerging field shows great promise for developing new therapeutic interventions to prevent and/or delay neurodegenerative disorders.
ACKNOWLEDGEMENTS
Declared none.
LIST OF ABBREVIATIONS
- AD
Alzheimer’s Disease
- PD
Parkinson’s Disease
- ROS
Reactive Oxygen Species
- RNS
Reactive Nitrogen Species
- CNS
Central Nervous System
- GPX
Glutathione Peroxidase
- SOD
Superoxide Dismutase
- GSH
Glutathione Reductase
- MAO
Monoamine Oxidase
- AChE
Acetylcholinesterase
- BChE
Butyrylcholinesterase
- TNF-α
Tumor Necrosis Factor α
- iNOS
Induced Nitric Oxide Synthase
- LPS
Lipopolysaccharides
AUTHORS’ CONTRIBUTIONS
The authors confirm their contribution to the paper as follows: Conceptualization S.P. and S.M.; Writing - Original Draft S.P., A.P., T.S., G.C., V.T., V.L. and S.M.; Writing - review & editing; S.P., A.P., T.S., V.T. and S.M.; Supervision S.P. and S.M. All authors reviewed the results and approved the final version of the manuscript.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This research was funded by “Ricerca Scientifica di Ateneo” (RSA) Grants (2023-2024) from University “Politecnica delle Marche” to Simona Magi and “Ministero dell’Istruzione and dell’ Università e della Ricerca” (MIUR) to Vincenzo Lariccia (PRIN 2022, grant no. 2022ZTKFXS).
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- 1.Dugger B.N., Dickson D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017;9(7):a028035. doi: 10.1101/cshperspect.a028035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.2022 Alzheimer’s disease facts and figures. Alzheimers Dement. 2022;18(4):700–789. doi: 10.1002/alz.12638. [DOI] [PubMed] [Google Scholar]
- 3.Mhyre T.R., Boyd J.T., Hamill R.W., Maguire-Zeiss K.A. Parkinson’s disease. Subcell. Biochem. 2012;65:389–455. doi: 10.1007/978-94-007-5416-4_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Inamdar N., Arulmozhi D., Tandon A., Bodhankar S. Parkinson’s disease: Genetics and beyond. Curr. Neuropharmacol. 2007;5(2):99–113. doi: 10.2174/157015907780866893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Klemmensen M.M., Borrowman S.H., Pearce C., Pyles B., Chandra B. Mitochondrial dysfunction in neurodegenerative disorders. Neurotherapeutics. 2024;21(1):e00292. doi: 10.1016/j.neurot.2023.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Butterfield D.A., Halliwell B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 2019;20(3):148–160. doi: 10.1038/s41583-019-0132-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kumar A., Sidhu J., Lui F., Tsao J.W. StatPearls. Treasure Island, FL: StatPearls; 2024. Alzheimer disease. [Google Scholar]
- 8.Chen Y., Yu Y. Tau and neuroinflammation in Alzheimer’s disease: Interplay mechanisms and clinical translation. J. Neuroinflammation. 2023;20(1):165. doi: 10.1186/s12974-023-02853-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tolar M., Hey J., Power A., Abushakra S. Neurotoxic soluble amyloid oligomers drive Alzheimer’s pathogenesis and represent a clinically validated target for slowing disease progression. Int. J. Mol. Sci. 2021;22(12):6355. doi: 10.3390/ijms22126355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gómez-Benito M., Granado N., García-Sanz P., Michel A., Dumoulin M., Moratalla R. Modeling Parkinson’s disease with the alpha-synuclein protein. Front. Pharmacol. 2020;11:356. doi: 10.3389/fphar.2020.00356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Freire M.A.M., Santos J.R. Parkinson’s disease: General features, effects of levodopa treatment and future directions. Front. Neuroanat. 2010;4:146. doi: 10.3389/fnana.2010.00146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mohd Sairazi N.S., Sirajudeen K.N.S. Natural products and their bioactive compounds: Neuroprotective potentials against neurodegenerative diseases. Evid. Based Complement. Alternat. Med. 2020;2020(1):6565396. doi: 10.1155/2020/6565396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sharifi-Rad M., Lankatillake C., Dias D.A., Docea A.O., Mahomoodally M.F., Lobine D., Chazot P.L., Kurt B., Boyunegmez T.T., Catarina M.A., Sharopov F., Martorell M., Martins N., Cho W.C., Calina D., Sharifi-Rad J. Impact of natural compounds on neurodegenerative disorders: From preclinical to pharmacotherapeutics. J. Clin. Med. 2020;9(4):1061. doi: 10.3390/jcm9041061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chen X., Guo C., Kong J. Oxidative stress in neurodegenerative diseases. Neural Regen. Res. 2012;7(5):376–385. doi: 10.3969/j.issn.1673-5374.2012.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jomova K., Raptova R., Alomar S.Y., Alwasel S.H., Nepovimova E., Kuca K., Valko M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023;97(10):2499–2574. doi: 10.1007/s00204-023-03562-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jones D.P. Radical-free biology of oxidative stress. Am. J. Physiol. Cell Physiol. 2008;295(4):C849–C868. doi: 10.1152/ajpcell.00283.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Piccirillo S., Magi S., Preziuso A., Serfilippi T., Cerqueni G., Orciani M., Amoroso S., Lariccia V. The hidden notes of redox balance in neurodegenerative diseases. Antioxidants. 2022;11(8):1456. doi: 10.3390/antiox11081456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Garbarino V.R., Orr M.E., Rodriguez K.A., Buffenstein R. Mechanisms of oxidative stress resistance in the brain: Lessons learned from hypoxia tolerant extremophilic vertebrates. Arch. Biochem. Biophys. 2015;576:8–16. doi: 10.1016/j.abb.2015.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Olufunmilayo E.O., Gerke-Duncan M.B., Holsinger R.M.D. Oxidative stress and antioxidants in neurodegenerative disorders. Antioxidants. 2023;12(2):517. doi: 10.3390/antiox12020517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Butterfield D.A. Perspectives on oxidative stress in Alzheimer’s disease and predictions of future research emphases. J. Alzheimers Dis. 2018;64(s1):S469–S479. doi: 10.3233/JAD-179912. [DOI] [PubMed] [Google Scholar]
- 21.Jiang T., Sun Q., Chen S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Prog. Neurobiol. 2016;147:1–19. doi: 10.1016/j.pneurobio.2016.07.005. [DOI] [PubMed] [Google Scholar]
- 22.Santos J.R., Gois A.M., Mendonça D.M., Freire M.A. Nutritional status, oxidative stress and dementia: The role of selenium in Alzheimer’s disease. Front. Aging Neurosci. 2014;6:206. doi: 10.3389/fnagi.2014.00206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Guo C., Sun L., Chen X., Zhang D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen. Res. 2013;8(21):2003–2014. doi: 10.3969/j.issn.1673-5374.2013.21.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Misrani A., Tabassum S., Yang L. Mitochondrial dysfunction and oxidative stress in Alzheimer’s disease. Front. Aging Neurosci. 2021;13:617588. doi: 10.3389/fnagi.2021.617588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Piccirillo S., Preziuso A., Amoroso S., Serfilippi T., Miceli F., Magi S., Lariccia V. A new K+channel-independent mechanism is involved in the antioxidant effect of XE-991 in an in vitro model of glucose metabolism impairment: Implications for Alzheimer’s disease. Cell Death Discov. 2022;8(1):391. doi: 10.1038/s41420-022-01187-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Piccirillo S., Magi S., Preziuso A., Castaldo P., Amoroso S., Lariccia V. Gateways for glutamate neuroprotection in Parkinson’s disease (PD): Essential role of EAAT3 and NCX1 revealed in an in vitro model of PD. Cells. 2020;9(9):2037. doi: 10.3390/cells9092037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Clemente-Suárez V., Redondo-Flórez L., Beltrán-Velasco A., Ramos-Campo D., Belinchón-deMiguel P., Martinez-Guardado I., Dalamitros A., Yáñez-Sepúlveda R., Martín-Rodríguez A., Tornero-Aguilera J. Mitochondria and brain disease: A comprehensive review of pathological mechanisms and therapeutic opportunities. Biomedicines. 2023;11(9):2488. doi: 10.3390/biomedicines11092488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Eckert A., Keil U., Marques C.A., Bonert A., Frey C., Schüssel K., Müller W.E. Mitochondrial dysfunction, apoptotic cell death, and Alzheimer’s disease. Biochem. Pharmacol. 2003;66(8):1627–1634. doi: 10.1016/S0006-2952(03)00534-3. [DOI] [PubMed] [Google Scholar]
- 29.Streit W.J., Mrak R.E., Griffin W.S.T. Microglia and neuroinflammation: A pathological perspective. J. Neuroinflammation. 2004;1(1):14. doi: 10.1186/1742-2094-1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yan Y-Q., Ma C-G., Ding Z-B., Song L-J., Wang Q., Kumar G. Astrocytes: A double-edged sword in neurodegenerative diseases. Neural Regen. Res. 2021;16(9):1702–1710. doi: 10.4103/1673-5374.306064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wang W.Y., Tan M.S., Yu J.T., Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015;3(10):136. doi: 10.3978/j.issn.2305-5839.2015.03.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rather M.A., Khan A., Alshahrani S., Rashid H., Qadri M., Rashid S., Alsaffar R.M., Kamal M.A., Rehman M.U. Inflammation and Alzheimer’s disease: Mechanisms and therapeutic implications by natural products. Mediators Inflamm. 2021;2021:1–21. doi: 10.1155/2021/9982954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Arena G., Sharma K., Agyeah G., Krüger R., Grünewald A., Fitzgerald J.C. Neurodegeneration and neuroinflammation in parkinson’s disease: A self-sustained loop. Curr. Neurol. Neurosci. Rep. 2022;22(8):427–440. doi: 10.1007/s11910-022-01207-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Adamu A., Li S., Gao F., Xue G. The role of neuroinflammation in neurodegenerative diseases: Current understanding and future therapeutic targets. Front. Aging Neurosci. 2024;16:1347987. doi: 10.3389/fnagi.2024.1347987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Riaz M., Khalid R., Afzal M., Anjum F., Fatima H., Zia S., Rasool G., Egbuna C., Mtewa A.G., Uche C.Z., Aslam M.A. Phytobioactive compounds as therapeutic agents for human diseases: A review. Food Sci. Nutr. 2023;11(6):2500–2529. doi: 10.1002/fsn3.3308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mutha R.E., Tatiya A.U., Surana S.J. Flavonoids as natural phenolic compounds and their role in therapeutics: An overview. Future J. Pharm. Sci. 2021;7(1):25. doi: 10.1186/s43094-020-00161-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Stagos D. Antioxidant activity of polyphenolic plant extracts. Antioxidants. 2019;9(1):19. doi: 10.3390/antiox9010019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rudrapal M., Khairnar S.J., Khan J., Dukhyil A.B., Ansari M.A., Alomary M.N., Alshabrmi F.M., Palai S., Deb P.K., Devi R. Dietary polyphenols and their role in oxidative stress-induced human diseases: Insights into protective effects, antioxidant potentials and mechanism(s) of action. Front. Pharmacol. 2022;13:806470. doi: 10.3389/fphar.2022.806470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Waheed Janabi A.H., Kamboh A.A., Saeed M., Xiaoyu L., BiBi J., Majeed F., Naveed M., Mughal M.J., Korejo N.A., Kamboh R., Alagawany M., Lv H. Flavonoid-rich foods (FRF): A promising nutraceutical approach against lifespan-shortening diseases. Iran. J. Basic Med. Sci. 2020;23(2):140–153. doi: 10.22038/IJBMS.2019.35125.8353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ullah A., Munir S., Badshah S.L., Khan N., Ghani L., Poulson B.G., Emwas A.H., Jaremko M. Important flavonoids and their role as a therapeutic agent. Molecules. 2020;25(22):5243. doi: 10.3390/molecules25225243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Devi S., Kumar V., Singh S.K., Dubey A.K., Kim J.J. Flavonoids: Potential candidates for the treatment of neurodegenerative disorders. Biomedicines. 2021;9(2):99. doi: 10.3390/biomedicines9020099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.de Andrade Teles R.B., Diniz T.C., Costa Pinto T.C., de Oliveira Júnior R.G., Gama e Silva M., de Lavor É.M., Fernandes A.W.C., de Oliveira A.P., de Almeida Ribeiro F.P.R., da Silva A.A.M., Cavalcante T.C.F., Quintans Júnior L.J., da Silva Almeida J.R.G. Flavonoids as therapeutic agents in Alzheimer’s and Parkinson’s diseases: A systematic review of preclinical evidences. Oxid. Med. Cell. Longev. 2018;2018(1):7043213. doi: 10.1155/2018/7043213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Meng-zhen S., Ju L., Lan-chun Z., Cai-feng D., Shu-da Y., Hao-fei Y., Wei-yan H. Potential therapeutic use of plant flavonoids in AD and PD. Heliyon. 2022;8(11):e11440. doi: 10.1016/j.heliyon.2022.e11440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Spencer J.P.E. Flavonoids and brain health: Multiple effects underpinned by common mechanisms. Genes Nutr. 2009;4(4):243–250. doi: 10.1007/s12263-009-0136-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pandey K.B., Rizvi S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009;2(5):270–278. doi: 10.4161/oxim.2.5.9498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Nájera-Maldonado J.M., Salazar R., Alvarez-Fitz P., Acevedo-Quiroz M., Flores-Alfaro E., Hernández-Sotelo D., Espinoza-Rojo M., Ramírez M. Phenolic compounds of therapeutic interest in neuroprotection. J. Xenobiot. 2024;14(1):227–246. doi: 10.3390/jox14010014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Grabska-Kobyłecka I., Szpakowski P., Król A., Książek-Winiarek D., Kobyłecki A., Głąbiński A., Nowak D. Polyphenols and their impact on the prevention of neurodegenerative diseases and development. Nutrients. 2023;15(15):3454. doi: 10.3390/nu15153454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ono K., Li L., Takamura Y., Yoshiike Y., Zhu L., Han F., Mao X., Ikeda T., Takasaki J., Nishijo H., Takashima A., Teplow D.B., Zagorski M.G., Yamada M. Phenolic compounds prevent amyloid β-protein oligomerization and synaptic dysfunction by site-specific binding. J. Biol. Chem. 2012;287(18):14631–14643. doi: 10.1074/jbc.M111.325456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Nakajima A., Ohizumi Y. Potential benefits of nobiletin, a citrus flavonoid, against Alzheimer’s disease and Parkinson’s disease. Int. J. Mol. Sci. 2019;20(14):3380. doi: 10.3390/ijms20143380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Maher P. Protective effects of fisetin and other berry flavonoids in Parkinson’s disease. Food Funct. 2017;8(9):3033–3042. doi: 10.1039/C7FO00809K. [DOI] [PubMed] [Google Scholar]
- 51.Cimini A., Gentile R., D’Angelo B., Benedetti E., Cristiano L., Avantaggiati M.L., Giordano A., Ferri C., Desideri G. Cocoa powder triggers neuroprotective and preventive effects in a human Alzheimer’s disease model by modulating BDNF signaling pathway. J. Cell. Biochem. 2013;114(10):2209–2220. doi: 10.1002/jcb.24548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Corti R., Flammer A.J., Hollenberg N.K., Lüscher T.F. Cocoa and cardiovascular health. Circulation. 2009;119(10):1433–1441. doi: 10.1161/CIRCULATIONAHA.108.827022. [DOI] [PubMed] [Google Scholar]
- 53.Andújar I., Recio M.C., Giner R.M., Ríos J.L. Cocoa polyphenols and their potential benefits for human health. Oxid. Med. Cell. Longev. 2012;2012:1–23. doi: 10.1155/2012/906252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Boncan D.A.T., Tsang S.S.K., Li C., Lee I.H.T., Lam H.M., Chan T.F., Hui J.H.L. Terpenes and terpenoids in plants: Interactions with environment and insects. Int. J. Mol. Sci. 2020;21(19):7382. doi: 10.3390/ijms21197382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yoo K.Y., Park S.Y. Terpenoids as potential anti-Alzheimer’s disease therapeutics. Molecules. 2012;17(3):3524–3538. doi: 10.3390/molecules17033524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Balakrishnan R., Azam S., Cho D.Y., Su-Kim I., Choi D.K. Natural phytochemicals as novel therapeutic strategies to prevent and treat Parkinson’s disease: Current knowledge and future perspectives. Oxid. Med. Cell. Longev. 2021;2021(1):6680935. doi: 10.1155/2021/6680935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lai Shi Min S., Liew S.Y., Chear N.J.Y., Goh B.H., Tan W.N., Khaw K.Y. Plant terpenoids as the promising source of cholinesterase inhibitors for anti-AD therapy. Biology (Basel) 2022;11(2):307. doi: 10.3390/biology11020307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chang W.H., Hsu H.T., Lin C.C., An L.M., Lee C.H., Ko H.H., Lin C.L., Lo Y.C. Linalool, a fragrance compound in plants, protects dopaminergic neurons and improves motor function and skeletal muscle strength in experimental models of Parkinson’s disease. Int. J. Mol. Sci. 2024;25(5):2514. doi: 10.3390/ijms25052514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mony T.J., Elahi F., Choi J.W., Park S.J. Neuropharmacological effects of terpenoids on preclinical animal models of psychiatric disorders: A review. Antioxidants. 2022;11(9):1834. doi: 10.3390/antiox11091834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chen C.C., Lee H.C., Chang J.H., Chen S.S., Li T.C., Tsai C.H., Cho D.Y., Hsieh C.L. Chinese herb astragalus membranaceus enhances recovery of hemorrhagic stroke: Double-blind, placebo-controlled, randomized study. Evid. Based Complement. Alternat. Med. 2012;2012:708452. doi: 10.1155/2012/708452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Costa I.M., Lima F.O.V., Fernandes L.C.B., Norrara B., Neta F.I., Alves R.D., Cavalcanti J.R.L.P., Lucena E.E.S., Cavalcante J.S., Rego A.C.M., Filho I.A., Queiroz D.B., Freire M.A.M., Guzen F.P. Astragaloside IV supplementation promotes a neuroprotective effect in experimental models of neurological disorders: A systematic review. Curr. Neuropharmacol. 2019;17(7):648–665. doi: 10.2174/1570159X16666180911123341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Aryal B., Raut B.K., Bhattarai S., Bhandari S., Tandan P., Gyawali K., Sharma K., Ranabhat D., Thapa R., Aryal D., Ojha A., Devkota H.P., Parajuli N. Potential therapeutic applications of plant-derived alkaloids against inflammatory and neurodegenerative diseases. Evid. Based Complement. Alternat. Med. 2022;2022:1–18. doi: 10.1155/2022/7299778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yatoo M.I., Gopalakrishnan A., Saxena A., Parray O.R., Tufani N.A., Chakraborty S., Tiwari R., Dhama K., Iqbal H.M.N. Anti-inflammatory drugs and herbs with special emphasis on herbal medicines for countering inflammatory diseases and disorders - A review. Recent Pat. Inflamm. Allergy Drug Discov. 2018;12(1):39–58. doi: 10.2174/1872213X12666180115153635. [DOI] [PubMed] [Google Scholar]
- 64.Zhao L., Wang L., Di S., Xu Q., Ren Q., Chen S., Huang N., Jia D., Shen X. Steroidal alkaloid solanine A from Solanum nigrum Linn. exhibits anti-inflammatory activity in lipopolysaccharide/interferon γ-activated murine macrophages and animal models of inflammation. Biomed. Pharmacother. 2018;105:606–615. doi: 10.1016/j.biopha.2018.06.019. [DOI] [PubMed] [Google Scholar]
- 65.Hussain G., Rasul A., Anwar H., Aziz N., Razzaq A., Wei W., Ali M., Li J., Li X. Role of plant derived alkaloids and their mechanism in neurodegenerative disorders. Int. J. Biol. Sci. 2018;14(3):341–357. doi: 10.7150/ijbs.23247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Linda E. Fellows 1 EAB. 5-hydroxy-l-tryptophan, 5-hydroxytryptamine and l-tryptophan-5-hydroxylase in Griffonia simplicifolia. Phytochemistry. 1970;9(11):2389–2396. [Google Scholar]
- 67.Muszyńska B., Łojewski M., Rojowski J., Opoka W., Sułkowska-Ziaja K. Natural products of relevance in the prevention and supportive treatment of depression. Psychiatr. Pol. 2015;49(3):435–453. doi: 10.12740/PP/29367. [DOI] [PubMed] [Google Scholar]
- 68.Rodondi P.Y., Graz B., Bonvin E. Should we collaborate with alternative medicines? Rev. Med. Suisse. 2012;8(325):224–225. doi: 10.53738/REVMED.2012.8.325.0224. [DOI] [PubMed] [Google Scholar]
- 69.Rondanelli M., Opizzi A., Faliva M., Bucci M., Perna S. Relationship between the absorption of 5-hydroxytryptophan from an integrated diet, by means of Griffonia simplicifolia extract, and the effect on satiety in overweight females after oral spray administration. Eat. Weight Disord. 2012;17(1):e22–e28. doi: 10.3275/8165. [DOI] [PubMed] [Google Scholar]
- 70.Wang X.Z., Wu F.H., Qu W., Liang J.Y. A new β-carboline alkaloid from the seeds of Griffonia simplicifolia. Chin. J. Nat. Med. 2013;11(4):401–405. doi: 10.1016/S1875-5364(13)60059-X. [DOI] [PubMed] [Google Scholar]
- 71.Rolland K.G., Pandey H.S., Rostand O.M., Vini T., Diane K., Priyanka S., Joseph D.A., Seth P. Cytoprotective action of Griffonia simplicifolia (DC.) Baill. against the oxidative stress caused by hydrogen peroxide (H2O2) on neurons and astrocytes. GSC Biol. Pharm. Sci. 2018;5(2):6–12. doi: 10.30574/gscbps.2018.5.2.0093. [DOI] [Google Scholar]
- 72.Mannino G., Serio G., Gaglio R., Maffei M.E., Settanni L., Di Stefano V., Gentile C. Biological activity and metabolomics of Griffonia simplicifolia seeds extracted with different methodologies. Antioxidants. 2023;12(9):1709. doi: 10.3390/antiox12091709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Młynarczyk K., Walkowiak-Tomczak D., Łysiak G.P. Bioactive properties of Sambucus nigra L. as a functional ingredient for food and pharmaceutical industry. J. Funct. Foods. 2018;40:377–390. doi: 10.1016/j.jff.2017.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mikulic-Petkovsek M., Samoticha J., Eler K., Stampar F., Veberic R. Traditional elderflower beverages: A rich source of phenolic compounds with high antioxidant activity. J. Agric. Food Chem. 2015;63(5):1477–1487. doi: 10.1021/jf506005b. [DOI] [PubMed] [Google Scholar]
- 75.Charlebois D.B.P., Finn C.E., Thomas A.L. Elderberry: Botany, horticulture, potential. Hortic. Rev. (Am. Soc. Hortic. Sci.) 2010;37:213–280. [Google Scholar]
- 76.Uncini Manganelli R.E., Zaccaro L., Tomei P.E. Antiviral activity in vitro of Urtica dioica L., Parietaria diffusa M. et K. and Sambucus nigra L. J. Ethnopharmacol. 2005;98(3):323–327. doi: 10.1016/j.jep.2005.01.021. [DOI] [PubMed] [Google Scholar]
- 77.Izzo A.A., di Carlo G., Biscardi D., de Fusco R., Mascolo N., Borrelli F., Capasso F., Fasulo M.P., Autore G. Biological screening of Italian medicinal plants for antibacterial activity. Phytother. Res. 1995;9(4):281–286. doi: 10.1002/ptr.2650090410. [DOI] [Google Scholar]
- 78.Harokopakis E., Albzreh M.H., Haase E.M., Scannapieco F.A., Hajishengallis G. Inhibition of proinflammatory activities of major periodontal pathogens by aqueous extracts from elder flower (Sambucus nigra). J. Periodontol. 2006;77(2):271–279. doi: 10.1902/jop.2006.050232. [DOI] [PubMed] [Google Scholar]
- 79.Beaux D., Fleurentin J., Mortier F. Effect of extracts ofOrthosiphon stamineus benth, Hieracium pilosella L., Sambucus nigra L. and Arctostaphylos uva-ursi (L.) spreng. in rats. Phytother. Res. 1999;13(3):222–225. doi: 10.1002/(SICI)1099-1573(199905)13:3<222::AID-PTR447>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 80.Palomino O., García-Aguilar A., González A., Guillén C., Benito M., Goya L. Biological actions and molecular mechanisms of Sambucus nigra L. in neurodegeneration: A cell culture approach. Molecules. 2021;26(16):4829. doi: 10.3390/molecules26164829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Khadeer Ahamed M.B., Krishna V., Dandin C.J. In vitro antioxidant and in vivo prophylactic effects of two γ-lactones isolated from Grewia tiliaefolia against hepatotoxicity in carbon tetrachloride intoxicated rats. Eur. J. Pharmacol. 2010;631(1-3):42–52. doi: 10.1016/j.ejphar.2009.12.034. [DOI] [PubMed] [Google Scholar]
- 82.Sheeja Malar D., Beema Shafreen R., Karutha Pandian S., Pandima Devi K. Cholinesterase inhibitory, anti-amyloidogenic and neuroprotective effect of the medicinal plant Grewia tiliaefolia – An in vitro and in silico study. Pharm. Biol. 2017;55(1):381–393. doi: 10.1080/13880209.2016.1241811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rajput A., Sharma P., Kumar N., Kaur S., Arora S. Neuroprotective activity of novel phenanthrene derivative from Grewia tiliaefolia by in vitro and in silico studies. Sci. Rep. 2023;13(1):2444. doi: 10.1038/s41598-023-29446-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Butterfield D.A., Boyd-Kimball D. Mitochondrial oxidative and nitrosative stress and Alzheimer disease. Antioxidants. 2020;9(9):818. doi: 10.3390/antiox9090818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Fonseca L.R., Rodrigues R.A., Ramos A.S., da Cruz J.D., Ferreira J.L.P., Silva J.R.A., Amaral A.C.F. Herbal medicinal products from Passiflora for anxiety: An unexploited potential. Sci. World J. 2020;2020:1–18. doi: 10.1155/2020/6598434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lee J., Jung H.Y., Lee S.I., Choi J.H., Kim S.G. Effects of Passiflora incarnata Linnaeus on polysomnographic sleep parameters in subjects with insomnia disorder: A double-blind randomized placebo-controlled study. Int. Clin. Psychopharmacol. 2020;35(1):29–35. doi: 10.1097/YIC.0000000000000291. [DOI] [PubMed] [Google Scholar]
- 87.Janda K., Wojtkowska K., Jakubczyk K., Antoniewicz J., Skonieczna-Żydecka K. Passiflora incarnata in neuropsychiatric disorders - A systematic review. Nutrients. 2020;12(12):3894. doi: 10.3390/nu12123894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.dos Reis Izolan L., da Silva D.M., Oliveira H.B.L., de Oliveira Salomon J.L., Peruzzi C.P., Garcia S.C., Dallegrave E., Zanotto C., Elisabetsky E., Gonçalves C.A., Arbo M.D., Konrath E.L., Leal M.B. Sintocalmy, a Passiflora incarnata based herbal, attenuates morphine withdrawal in mice. Neurochem. Res. 2021;46(5):1092–1100. doi: 10.1007/s11064-021-03237-w. [DOI] [PubMed] [Google Scholar]
- 89.Miyasaka L.S., Atallah A.N., Soares B.G. Passiflora for anxiety disorder. Cochrane Database Syst. Rev. 2007;(1):CD004518. doi: 10.1002/14651858.CD004518.pub2. [DOI] [PubMed] [Google Scholar]
- 90.Ingale S., Kasture S. Protective effect of standardized extract of Passiflora incarnata flower in Parkinson’s and Alzheimer’s disease. Anc. Sci. Life. 2017;36(4):200–206. doi: 10.4103/asl.ASL_231_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gadioli I.L., da Cunha M.S.B., de Carvalho M.V.O., Costa A.M., Pineli L.L.O. A systematic review on phenolic compounds in Passiflora plants: Exploring biodiversity for food, nutrition, and popular medicine. Crit. Rev. Food Sci. Nutr. 2018;58(5):785–807. doi: 10.1080/10408398.2016.1224805. [DOI] [PubMed] [Google Scholar]
- 92.Kim G.H., Lim K., Yang H.S., Lee J.K., Kim Y., Park S.K., Kim S.H., Park S., Kim T.H., Moon J.S., Hwang I.K., Yoon Y.S., Seo H.S., Nam S.M., Kim M.Y., Yoon S.G., Seong J.K., Yi S.S. Improvement in neurogenesis and memory function by administration of Passiflora incarnata L. extract applied to sleep disorder in rodent models. J. Chem. Neuroanat. 2019;98:27–40. doi: 10.1016/j.jchemneu.2019.03.005. [DOI] [PubMed] [Google Scholar]
- 93.Blecharz-Klin K., Pyrzanowska J., Piechal A., Joniec-Maciejak I., Wawer A., Jawna-Zboińska K., Mirowska-Guzel D., Widy-Tyszkiewicz E. Effect of Passiflora incarnata L. extract on exploratory behaviour and neurotransmitters level in structures involved in motor functions in rats. J. Preclin. Clin. Res. 2024;18(1):1–10. doi: 10.26444/jpccr/178182. [DOI] [Google Scholar]
- 94.Martinelli F., Perrone A., Yousefi S., Papini A., Castiglione S., Guarino F., Cicatelli A., Aelaei M., Arad N., Gholami M., Salami S. Botanical, phytochemical, anti-microbial and pharmaceutical characteristics of hawthorn (Crataegus monogyna Jacq.), rosaceae. Molecules. 2021;26(23):7266. doi: 10.3390/molecules26237266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Cui T., Nakamura K., Tian S., Kayahara H., Tian Y.L. Polyphenolic content and physiological activities of Chinese hawthorn extracts. Biosci. Biotechnol. Biochem. 2006;70(12):2948–2956. doi: 10.1271/bbb.60361. [DOI] [PubMed] [Google Scholar]
- 96.Schüssler M., Hölzl J., Fricke U. Myocardial effects of flavonoids from Crataegus species. Arzneimittelforschung. 1995;45(8):842–845. [PubMed] [Google Scholar]
- 97.Weihmayr T., Ernst E. Therapeutic effectiveness of Crataegus. Fortschr. Med. 1996;114(1-2):27–29. [PubMed] [Google Scholar]
- 98.Rashid D.P.J. Antioxidant and antiradical activities assessment in two hawthorn species fruit components. Curr. Nutr. Food Sci. 2013;9(1):52–58. [Google Scholar]
- 99.Ali M., Muhammad S., Shah M.R., Khan A., Rashid U., Farooq U., Ullah F., Sadiq A., Ayaz M., Ali M., Ahmad M., Latif A. Neurologically potent molecules from Crataegus oxyacantha; Isolation, anticholinesterase inhibition, and molecular docking. Front. Pharmacol. 2017;8:327. doi: 10.3389/fphar.2017.00327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kostić D.A., Velicković J.M., Mitić S.S., Mitić M.N., Randelović S.S. Phenolic content, and antioxidant and antimicrobial activities of Crataegus oxyacantha L. (Rosaceae) fruit extract from Southeast Serbia. Trop. J. Pharm. Res. 2012;11(1):117–124. doi: 10.4314/tjpr.v11i1.15. [DOI] [Google Scholar]
- 101.Saoudi M., Slama-Ben Salem R.B., Salem M.B., Brahmi N., Badraoui R., Nasri M., El Feki A. Beneficial effects of crataegus oxyacantha extract on neurobehavioral deficits and brain tissue damages induced by an insecticide mixture of deltamethrin and chlorpyrifos in adult wistar rats. Biomed. Pharmacother. 2019;114:108795. doi: 10.1016/j.biopha.2019.108795. [DOI] [PubMed] [Google Scholar]
- 102.Paul S., Sharma S., Paliwal S.K., Kasture S. Role of Crataegus oxyacantha (Hawthorn) on scopolamine induced memory deficit and monoamine mediated behaviour in rats. Orient. Pharm. Exp. Med. 2017;17(4):315–324. doi: 10.1007/s13596-017-0273-y. [DOI] [Google Scholar]
- 103.Lanje C.N. Medicinal natural drug of Valerian (Valerina officinalis): An-over review. Am. J. PharmTech. Res. 2020;10(1):149–172. [Google Scholar]
- 104.Sundaresan Nandhini K.B.N., Kaliappan I. Valeriana officinalis: A review of its traditional uses, phytochemistry and pharmacology. Asian J. Pharm. Clin. Res. 2018;11(1):36. [Google Scholar]
- 105.Das G., Shin H.S., Tundis R., Gonçalves S., Tantengco O.A.G., Campos M.G., Acquaviva R., Malfa G.A., Romano A., Robles J.A.H., Clores M.Q., Patra J.K. Plant species of sub-family valerianaceae - A review on its effect on the central nervous system. Plants. 2021;10(5):846. doi: 10.3390/plants10050846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Küpeli A.E., Tatlı Ç.I., Şeker K.G., Carpar E., Sobarzo-Sánchez E., Capasso R. Natural compounds as medical strategies in the prevention and treatment of psychiatric disorders seen in neurological diseases. Front. Pharmacol. 2021;12:669638. doi: 10.3389/fphar.2021.669638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Patočka J., Jakl J. Biomedically relevant chemical constituents of Valeriana officinalis. J. Appl. Biomed. 2010;8(1):11–18. doi: 10.2478/v10136-009-0002-z. [DOI] [Google Scholar]
- 108.Grosso C., Valentão P., Ferreres F., Andrade P. The use of flavonoids in central nervous system disorders. Curr. Med. Chem. 2013;20(37):4694–4719. doi: 10.2174/09298673113209990155. [DOI] [PubMed] [Google Scholar]
- 109.de Oliveria D.M., Barreto G., De Andrade D.V.G., Saraceno E., Aon-Bertolino L., Capani F., Dos Santos El Bachá R., Giraldez L.D. Cytoprotective effect of Valeriana officinalis extract on an in vitro experimental model of Parkinson disease. Neurochem. Res. 2009;34(2):215–220. doi: 10.1007/s11064-008-9749-y. [DOI] [PubMed] [Google Scholar]
- 110.Malva J.O., Santos S., Macedo T. Neuroprotective properties of Valeriana officinalis extracts. Neurotox. Res. 2004;6(2):131–140. doi: 10.1007/BF03033215. [DOI] [PubMed] [Google Scholar]
- 111.Marawne H., Mohammadhassan R., Mohammadalipour Z. Valerian (Valeriana officinalis) extract inhibits TNF-α and iNOS gene expression in mouse LPS-activated microglial cells. Trad. Med. Res. 2022;7(5):47. doi: 10.53388/TMR20220320003. [DOI] [Google Scholar]
- 112.Lee Y., Jung J.C., Jang S., Kim J., Ali Z., Khan I.A., Oh S. Anti-inflammatory and neuroprotective effects of constituents isolated from Rhodiola rosea. Evid. Based Complement. Alternat. Med. 2013;2013:514049. doi: 10.1155/2013/514049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhuang W., Yue L., Dang X., Chen F., Gong Y., Lin X., Luo Y. Rosenroot (Rhodiola): Potential applications in aging-related diseases. Aging Dis. 2019;10(1):134–146. doi: 10.14336/AD.2018.0511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Qu Z.Q., Zhou Y., Zeng Y.S., Li Y., Chung P. Pretreatment with Rhodiola rosea extract reduces cognitive impairment induced by intracerebroventricular streptozotocin in rats: Implication of anti-oxidative and neuroprotective effects. Biomed. Environ. Sci. 2009;22(4):318–326. doi: 10.1016/S0895-3988(09)60062-3. [DOI] [PubMed] [Google Scholar]
- 115.Bocquet L., Sahpaz S., Hilbert J.L., Rambaud C., Rivière C. Humulus lupulus L., a very popular beer ingredient and medicinal plant: Overview of its phytochemistry, its bioactivity, and its biotechnology. Phytochem. Rev. 2018;17(5):1047–1090. doi: 10.1007/s11101-018-9584-y. [DOI] [Google Scholar]
- 116.Yen T.L., Hsu C.K., Lu W.J., Hsieh C.Y., Hsiao G., Chou D.S., Wu G.J., Sheu J.R. Neuroprotective effects of xanthohumol, a prenylated flavonoid from hops (Humulus lupulus), in ischemic stroke of rats. J. Agric. Food Chem. 2012;60(8):1937–1944. doi: 10.1021/jf204909p. [DOI] [PubMed] [Google Scholar]
- 117.Sasaoka N., Sakamoto M., Kanemori S., Kan M., Tsukano C., Takemoto Y., Kakizuka A. Long-term oral administration of hop flower extracts mitigates Alzheimer phenotypes in mice. PLoS One. 2014;9(1):e87185. doi: 10.1371/journal.pone.0087185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Palmioli A., Mazzoni V., De Luigi A., Bruzzone C., Sala G., Colombo L., Bazzini C., Zoia C.P., Inserra M., Salmona M., De Noni I., Ferrarese C., Diomede L., Airoldi C. Alzheimer’s disease prevention through natural compounds: Cell-free, in vitro, and in vivo dissection of hop (Humulus lupulus L.) multitarget activity. ACS Chem. Neurosci. 2022;13(22):3152–3167. doi: 10.1021/acschemneuro.2c00444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Tung M.C., Fung K.M., Hsu H.M., Tseng T.S. Discovery of 8-prenylnaringenin from hop ( Humulus lupulus L.) as a potent monoacylglycerol lipase inhibitor for treatments of neuroinflammation and Alzheimer’s disease. RSC Advances. 2021;11(49):31062–31072. doi: 10.1039/D1RA05311F. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.El Mihyaoui A., Esteves da Silva J.C.G., Charfi S., Candela C.M.E., Lamarti A., Arnao M.B. Chamomile (Matricaria chamomilla L.): A review of ethnomedicinal use, phytochemistry and pharmacological uses. Life (Basel) 2022;12(4):479. doi: 10.3390/life12040479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Dai Y.L., Li Y., Wang Q., Niu F.J., Li K.W., Wang Y.Y., Wang J., Zhou C.Z., Gao L.N. Chamomile: A review of its traditional uses, chemical constituents, pharmacological activities and quality control studies. Molecules. 2022;28(1):133. doi: 10.3390/molecules28010133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Saeedi M.K.M., Shahsavari K., Manayi A. Matricaria chamomilla: An updated review on biological activities of the plant and constituents. Res. J. Pharmacogn. 2023;11(1):109–136. [Google Scholar]
- 123.Sah A., Naseef P.P., Kuruniyan M.S., Jain G.K., Zakir F., Aggarwal G. A comprehensive study of therapeutic applications of chamomile. Pharmaceuticals (Basel) 2022;15(10):1284. doi: 10.3390/ph15101284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Alahmady N.F., Alkhulaifi F.M., Abdullah M.M., Ali Alharbi A., Allohibi A., Alsubhi N.H., Ahmed A.W. Biochemical characterization of chamomile essential oil: Antioxidant, antibacterial, anticancer and neuroprotective activity and potential treatment for Alzheimer’s disease. Saudi J. Biol. Sci. 2024;31(2):103912. doi: 10.1016/j.sjbs.2023.103912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ionita R., Postu P.A., Mihasan M., Gorgan D.L., Hancianu M., Cioanca O., Hritcu L. Ameliorative effects of Matricaria chamomilla L. hydroalcoholic extract on scopolamine-induced memory impairment in rats: A behavioral and molecular study. Phytomedicine. 2018;47:113–120. doi: 10.1016/j.phymed.2018.04.049. [DOI] [PubMed] [Google Scholar]
- 126.Shakeri A., Sahebkar A., Javadi B. Melissa officinalis L. – A review of its traditional uses, phytochemistry and pharmacology. J. Ethnopharmacol. 2016;188:204–228. doi: 10.1016/j.jep.2016.05.010. [DOI] [PubMed] [Google Scholar]
- 127.Sipos S., Moacă E.A., Pavel I.Z., Avram Ş., Crețu O.M., Coricovac D., Racoviceanu R.M., Ghiulai R., Pană R.D., Şoica C.M., Borcan F., Dehelean C.A., Crăiniceanu Z. Melissa officinalis L. aqueous extract exerts antioxidant and antiangiogenic effects and improves physiological skin parameters. Molecules. 2021;26(8):2369. doi: 10.3390/molecules26082369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Patora J., Klimek B. Flavonoids from lemon balm (Melissa officinalis L., Lamiaceae). Acta Pol. Pharm. 2002;59(2):139–143. [PubMed] [Google Scholar]
- 129.Draginic N., Andjic M., Jeremic J., Zivkovic V., Kocovic A., Tomovic M., Bozin B., Kladar N., Bolevich S., Jakovljevic V., Milosavljevic I. Anti-inflammatory and antioxidant effects of Melissa officinalis extracts: A comparative study. Iran. J. Pharm. Res. 2022;21(1):e126561. doi: 10.5812/ijpr-126561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ozarowski M., Mikolajczak P.L., Piasecka A., Kachlicki P., Kujawski R., Bogacz A., Bartkowiak-Wieczorek J., Szulc M., Kaminska E., Kujawska M., Jodynis-Liebert J., Gryszczynska A., Opala B., Lowicki Z., Seremak-Mrozikiewicz A., Czerny B. Influence of the Melissa officinalis leaf extract on long‐term memory in scopolamine animal model with assessment of mechanism of action. Evid. Based Complement. Alternat. Med. 2016;2016(1):9729818. doi: 10.1155/2016/9729818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Akhondzadeh S., Noroozian M., Mohammadi M., Ohadinia S., Jamshidi A.H., Khani M. Melissa officinalis extract in the treatment of patients with mild to moderate Alzheimer’s disease: A double blind, randomised, placebo controlled trial. J. Neurol. Neurosurg. Psychiatry. 2003;74(7):863–866. doi: 10.1136/jnnp.74.7.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Martins E.N., Pessano N.T.C., Leal L., Roos D.H., Folmer V., Puntel G.O., Rocha J.B.T., Aschner M., Ávila D.S., Puntel R.L. Protective effect of Melissa officinalis aqueous extract against Mn-induced oxidative stress in chronically exposed mice. Brain Res. Bull. 2012;87(1):74–79. doi: 10.1016/j.brainresbull.2011.10.003. [DOI] [PubMed] [Google Scholar]
- 133.Ammon H., Wahl M. Pharmacology of Curcuma longa. Planta Med. 1991;57(1):1–7. doi: 10.1055/s-2006-960004. [DOI] [PubMed] [Google Scholar]
- 134.Zielińska A., Alves H., Marques V., Durazzo A., Lucarini M., Alves T.F., Morsink M., Willemen N., Eder P., Chaud M.V., Severino P., Santini A., Souto E.B. Properties, extraction methods, and delivery systems for curcumin as a natural source of beneficial health effects. Medicina (Kaunas) 2020;56(7):336. doi: 10.3390/medicina56070336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Adami R., Bottai D. Curcumin and neurological diseases. Nutr. Neurosci. 2022;25(3):441–461. doi: 10.1080/1028415X.2020.1760531. [DOI] [PubMed] [Google Scholar]
- 136.Kim D.S.H.L., Park S.Y., Kim J.Y. Curcuminoids from Curcuma longa L. (Zingiberaceae) that protect PC12 rat pheochromocytoma and normal human umbilical vein endothelial cells from βA(1–42) insult. Neurosci. Lett. 2001;303(1):57–61. doi: 10.1016/S0304-3940(01)01677-9. [DOI] [PubMed] [Google Scholar]
- 137.Rajeswari A. Curcumin protects mouse brain from oxidative stress caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Eur. Rev. Med. Pharmacol. Sci. 2006;10(4):157–161. [PubMed] [Google Scholar]
- 138.Genchi G., Lauria G., Catalano A., Carocci A., Sinicropi M.S. Neuroprotective effects of curcumin in neurodegenerative diseases. Foods. 2024;13(11):1774. doi: 10.3390/foods13111774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Kundu P., Das M., Tripathy K., Sahoo S.K. Delivery of dual drug loaded lipid based nanoparticles across the blood–brain barrier impart enhanced neuroprotection in a rotenone induced mouse model of Parkinson’s disease. ACS Chem. Neurosci. 2016;7(12):1658–1670. doi: 10.1021/acschemneuro.6b00207. [DOI] [PubMed] [Google Scholar]
- 140.da Costa I.M., de Moura Freire M.A., de Paiva Cavalcanti J.R.L., de Araújo D.P., Norrara B., Moreira Rosa I.M.M., de Azevedo E.P., do Rego A.C.M., Filho I.A., Guzen F.P. Supplementation with Curcuma longa reverses neurotoxic and behavioral damage in models of alzheimer’s disease: A systematic review. Curr. Neuropharmacol. 2019;17(5):406–421. doi: 10.2174/0929867325666180117112610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Reddy P.H., Manczak M., Yin X., Grady M.C., Mitchell A., Kandimalla R., Kuruva C.S. Protective effects of a natural product, curcumin, against amyloid β induced mitochondrial and synaptic toxicities in Alzheimer’s disease. J. Investig. Med. 2016;64(8):1220–1234. doi: 10.1136/jim-2016-000240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zahra W., Birla H., Singh S.S., Rathore A.S., Dilnashin H., Singh R., Keshri P.K., Gautam P., Singh S.P. Neuroprotection by Mucuna pruriens in neurodegenerative diseases. Neurochem. Res. 2022;47(7):1816–1829. doi: 10.1007/s11064-022-03591-3. [DOI] [PubMed] [Google Scholar]
- 143.Dhanasekaran M., Tharakan B., Manyam B.V. Antiparkinson drug – Mucuna pruriens shows antioxidant and metal chelating activity. Phytother. Res. 2008;22(1):6–11. doi: 10.1002/ptr.2109. [DOI] [PubMed] [Google Scholar]
- 144.Misra L., Wagner H. Extraction of bioactive principles from Mucuna pruriens seeds. Indian J. Biochem. Biophys. 2007;44(1):56–60. [PubMed] [Google Scholar]
- 145.Manyam B.V., Dhanasekaran M., Hare T.A. Neuroprotective effects of the antiparkinson drug Mucuna pruriens. Phytother. Res. 2004;18(9):706–712. doi: 10.1002/ptr.1514. [DOI] [PubMed] [Google Scholar]
- 146.Guzen F.P., Neta F.I., Da Costa I.M., Lima F.O.V., Fernandes L.C.B., Cavalcanti J.R.L.D.P., Freire M.A.D.M., De Souza Lucena E.E., Meneses Do Rêgo A.C., Filho I.A., De Azevedo E.P. Effects of Mucuna pruriens (L.) supplementation on experimental models of parkinson’s disease: A systematic review. Pharmacogn. Rev. 2018;12(23):78–84. doi: 10.4103/phrev.phrev_46_17. [DOI] [Google Scholar]
- 147.Nayak V.S., Kumar N., D’Souza A.S., Nayak S.S., Cheruku S.P., Pai K.S.R. The effects of Mucuna pruriens extract on histopathological and biochemical features in the rat model of ischemia. Neuroreport. 2017;28(18):1195–1201. doi: 10.1097/WNR.0000000000000888. [DOI] [PubMed] [Google Scholar]
- 148.Ahmed S., Khan H., Mirzaei H., Hasan M.M., Eddouks M., Daglia M. Herbal drug interaction: Mechanistic details through pharmacokinetic portfolio. CNS Neurol. Disord. Drug Targets. 2021;20(8):677–686. doi: 10.2174/1871527319666201008151710. [DOI] [PubMed] [Google Scholar]
- 149.Abolarin P.O., Amin A., Nafiu A.B., Ogundele O.M., Owoyele B.V. Optimization of Parkinson’s disease therapy with plant extracts and nutrition’s evolving roles. IBRO Neurosci. Rep. 2024;17:1–12. doi: 10.1016/j.ibneur.2024.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Abebe W. Herbal medication: Potential for adverse interactions with analgesic drugs. J. Clin. Pharm. Ther. 2002;27(6):391–401. doi: 10.1046/j.1365-2710.2002.00444.x. [DOI] [PubMed] [Google Scholar]

