Next Article in Journal
Surface Activity, Wetting, and Aggregation of a Perfluoropolyether Quaternary Ammonium Salt Surfactant with a Hydroxyethyl Group
Previous Article in Journal
Physicochemical Characterization of Dextran HE29 Produced by the Leuconostoc citreum HE29 Isolated from Traditional Fermented Pickle
Previous Article in Special Issue
2-Alkyl-Substituted-4-Amino-Thieno[2,3-d]Pyrimidines: Anti-Proliferative Properties to In Vitro Breast Cancer Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of Phytochemical Potency as a Natural Anti-Helicobacter pylori and Neuroprotective Agent

1
Department of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung 40201, Taiwan
2
Faculty of Agricultural Technology, Widya Mandala Catholic University Surabaya, Surabaya 60265, Indonesia
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(20), 7150; https://doi.org/10.3390/molecules28207150
Submission received: 1 September 2023 / Revised: 13 October 2023 / Accepted: 13 October 2023 / Published: 18 October 2023

Abstract

:
Phytochemicals are plant secondary metabolites that show health benefits for humans due to their bioactivity. There is a huge variety of phytochemicals that have already been identified, and these compounds can act as antimicrobial and neuroprotection agents. Due to their anti-microbial activity and neuroprotection, several phytochemicals might have the potency to be used as natural therapeutic agents, especially for Helicobacter pylori infection and neurodegenerative disease, which have become a global health concern nowadays. According to previous research, there are some connections between H. pylori infection and neurodegenerative diseases, especially Alzheimer’s disease. Hence, this comprehensive review examines different kinds of phytochemicals from natural sources as potential therapeutic agents to reduce H. pylori infection and improve neurodegenerative disease. An additional large-scale study is needed to establish the connection between H. pylori infection and neurodegenerative disease and how phytochemicals could improve this condition.

1. Introduction

Helicobacter pylori infection is one of the global health problems. More than 50% of the population in the world is affected, mostly in developing countries [1]. H. pylori attaches to the human stomach; induces a change in gastric physiology; and is highly associated with gastric ulcers, which further progress into gastric cancer [2]. H. pylori can colonize and infect gastric tissue because of virulent factors such as urease, lipopolysaccharide (LPS), vacuolating cytotoxin A (VacA), cytotoxin-associated gene A (CagA), and some others [3]. Until now, the main treatment for H. pylori infection is to use the combination of two antibiotics together with a bismuth compound and/or antacid agent such proton pump inhibitor (PPI), which is called quadruple therapy and provides an eradication rate of more than 80% [4]. The usage of antibiotics in H. pylori offers another concern of some side effects as well as antibiotic resistance problems [5]. Recent studies show that H. pylori infection contributes to the progression of neurodegenerative diseases.
Neurodegenerative diseases (NDs) are disorders that affect the central nervous system and that are mostly caused by neuronal cell death, which causes impairment of the cognitive and motoric system [6]. There are many risk factors associated with ND progression, but its pathogenesis has still been unclear until now. Several diseases are classified as NDs such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD) [7]. These diseases have different characteristics, but most of them share the same hallmarks, which are neuronal cell death and neuroinflammation [8,9]. Until now, ND has been classified as an incurable disease, and medication might have a small impact on improving a patient’s condition [10]. Evidence of nutraceuticals on NDs is still deficient, in terms of whether together with normal medication, they could provide better effects on subjects with NDs.
There are several hypotheses about the possible connection between H. pylori infection and NDs. H. pylori affect the absorption of folate and vitamin B-12, which causes the elevation of homocysteine level and induces neurotoxicity. Furthermore, H. pylori cross the blood–brain barrier and induce amyloid deposition in the brain [11]. Another study showed that the outer membrane vesicles of H. pylori that were injected into mice altered astrocyte function and induced neuronal damage in the mouse brain [12]. In PD, it showed that H. pylori infection is related to the progression of the disease and increases the requirement of medication for PD [13]. This evidence might provide a clue about the connection between neurodegenerative disease and H. pylori infection.
Phytochemicals are secondary metabolites of plants, which are non-nutritive bio-active compounds synthesized for natural defenses of the plant against pests [14,15,16,17]. Phytochemicals found in fruits, vegetables, nuts, and grains provide health benefits. Many studies showed that phytochemicals from different natural sources act as antibacterial agents or neuroprotective agents [17,18]. Önem et al. showed that stalk extracts from two different cultivars of Prunus avium L. inhibited Gram-positive bacteria and reduced the biofilm formation of bacteria by up to 75% [19]. Li et al. showed that supplementation of proanthocyanidins (PAC)-rich cranberry juice (44 mg of PAC per portion) twice a day for 8 weeks significantly reduced H. pylori infection [20]. Desideri et al. reported that high intake (990 mg/day) of dietary flavonols from cocoa for 8 weeks significantly improved cognitive function in mild cognitive impairment subjects compared to those of low intake (45 mg/day) of cocoa flavonols [21]. Kent et al. pointed out that the intervention of anthocyanin-rich cherry juice for 12 weeks significantly improved verbal fluency and short-term and long-term memory in subjects with dementia [22]. Past studies showed that phytochemicals can be used as drug alternatives to treat H. pylori and neurodegenerative disease and reduce the risk of antibiotic resistance and complications due to the medication. Hence, this review discusses the potential of phytochemicals from various sources for H. pylori infection and also neuroprotection in in vitro and in vivo studies.

2. H. pylori

H. pylori is a Gram-negative spiral bacterium that is found in the human stomach and is associated with gastric ulcer and advanced gastric cancer [2,23,24]. The infection of H. pylori shows no symptoms in most cases, but it depends on the immune response of the individual and the severity of the syndrome. Most symptoms of H. pylori infection are correlated with the gastric ulcer and inflammation in the gastric tissue [25]. H. pylori is considered a special bacterium due to the virulence factors (Figure 1) that help to colonize in the human stomach, such as VacA, CagA, urease, LPS, and different kinds of adhesins [3].

2.1. VacA

VacA is one of the virulence factors possessed by H. pylori. VacA is the major toxic 88 kDa protein that is secreted from H. pylori through type V auto transport secretion system (T5SS), which binds to the host cell and causes vacuolation of the cell [26].
VacA plays an important role in the colonization of H. pylori in the gastric mucosa, stimulating the autophagy pathway in cells and disrupting lysosomal trafficking that causes the accumulation of dysfunctional autophagosomes and the formation of large intracellular vacuoles to promote the intracellular survival of H. pylori [27]. Furthermore, it induces different responses in infected cells such as vacuole formation, cytochrome c release, and forming channels in the mitochondria [28]. It also induces cell apoptosis because of increasing cytochrome c release from mitochondria. Cytochrome c combines with Apaf-1 and caspase-9 to stimulate the production of caspase-3 and caspase-7, resulting in cell apoptosis [29,30]. VacA can disrupt the tight junction to alter the tissue structure and increase the adhesion of H. pylori to epithelial cells [26,31,32].

2.2. CagA

CagA is a 120 to 145 kDa protein that can be injected into the host cell by using a type IV secretion system (T4SS) after the adhesion of H. pylori to the host cell [33]. H. pylori is divided into two different strains based on the presence of CagA: CagA-positive and CagA-negative strains. The cagA-positive strain is more virulent than the CagA-negative strain and is associated with higher gastric inflammation [34].
The effects of CagA on the host cell are independent of the phosphorylation process. The most noticeable is to disrupt the cell’s tight junction and induce cell morphology changes [35]. Non-phosphorylated CagA also can activate serum response elements further affect the cell cycle and induce inflammatory response [36].

2.3. Urease

Urease is a 550 kDa molecule consisting of UreA and UreB subunits. Urease plays a crucial role in the survival of H. pylori in the human stomach. H. pylori produces urease in acidic conditions, which breaks down urea and releases ammonia to neutralize the acidic condition in the human stomach [37]. pH increases in the stomach alter the protective mucous layer and also dysregulate the gastric epithelial cell tight junction [38].

2.4. Pathophysiology of H. pylori Infection

H. pylori infection is associated with chronic gastritis, gastric ulcers, and gastric cancer [1]. Development of gastric problems due to H. pylori infection is mostly caused by alteration of the gastric physiology and microenvironment, which induces an immune response from the human body [39]. This immune response is due to the activity of the H. pylori virulence factors such as CagA, VacA, and urease, and the response might be different depending on the age [40,41]. Immune response due to H. pylori infection is mediated by Toll-like receptors (TLRs) and microRNA, which can promote or suppress the immune response [42]. After reaching the stomach, H. pylori move to the mucous layer to evade the acid condition with the help of urease and attach to epithelial cells with the help of different kinds of adhesins such as BabA, SabA, AlpA/B, HopZ, and OipA [43]. After binding to the host cell, H. pylori inject different kinds of toxins such as CagA and VacA, depending on the strain, being able to induce inflammatory responses and upregulation of pro-inflammatory cytokines secretion [1].

2.5. Diagnosis and Treatment

There are various methods to identify and diagnose H. pylori. The invasive tests are based on gastric biopsy and peripheral samples to check the infection of H. pylori. On the other hand, the non-invasive method is to use the Urea Breath Test (UBT) C13 or C14 [1].
UBT is one of the most popular methods to diagnose H. pylori infection due to its high sensitivity and is considered the gold standard of the non-invasive method [1]. UBT is based on the reaction of C13-labeled urea and bacterial urease secreted from H. pylori, which produce ammonia (NH3) and C13-labeled carbon dioxide in the breath. The concentration of the C13 isotope is determined by using gas chromatography and considered positive if the Delta Over Baseline (DOB) value is ≥4‰ [44,45,46].
Treatment of H. pylori infection is usually conducted by using antibiotics and combination with PPI and/or with bismuth. Monotherapy (single antibiotic) was used in the past, but the efficacy was poor. The addition of PPI is used as dual therapy in some countries. Overuse of antibiotics induces the mutation and resistance of H. pylori and produces some side effects such as dizziness, vomiting, and allergy [47,48].

3. NDs

NDs are diseases that occurs in the central nervous system (CNS), being characterized by the progressive reduction of neuronal cells in the brain due to cell death [49]. Until now, there have been no medications that can cure these diseases due to the characteristic of neuronal cells, being unable to regenerate themselves after cell damage and death [50]. The diseases mostly affect elderly people aged >60 years. Recently, it become a public health concern due to them also affecting the younger generation worldwide [51,52,53]. In general, neurodegenerative diseases share a similar major hallmark, which is neuronal cell death, with the major pathways being apoptosis and necrosis with difference and chronic neuroinflammation [8,9]. These conditions could occur due to stress accumulation and misfolded protein deposits, which can induce cytotoxicity events such as impairment of cell signaling, DNA damage, mitochondrial dysfunction, and increased ROS production, which leads to neuronal cell death [8,49,54]. There are various manifestations of NDs, such as AD, PD, HD, and amyotrophic lateral sclerosis [10,50].

4. AD

AD is a neurodegenerative disease that causes a decline in cognitive functions and interferes with daily living activities. It is the most common form of dementia, especially for peoples aged over 65 [55]. In the United States, 1 out of 10 peoples at age over 65 years is estimated to suffer from AD, and the prevalence increases with age [56]. The major characteristics of the early stages of this disease are short-term memory loss, including impairment of problem-solving ability, multitasking, and abstract thinking problems. The later stage includes subsequent changes in cognitive ability and behavior [57]. Different stages are classified according to the cognitive impairment degree, including preclinical, mild, and dementia stages [55].
AD is a complicated disease. Its initiation and progression into dementia are associated with Aβ and NFT formation [58]. Aβ is a peptide, consisting of 42 amino acids derived from APP [59]. In a normal pathway (Figure 2), APP is cleaved by α-secretase activity producing a large soluble fraction called sAPPα and αCTF, which further cleaved by the activity of γ-secretase, producing AICD and a protein fragment called p83, which rapidly degraded [60]. Aβ is cleaved from APP by β-secretase, and γ-secretase by the amyloidogenic pathway (Figure 2), releasing C terminal peptides that tend to aggregate into oligomers and fibrils to form the senile plaque in the brain [61]. Aggregates of Aβ can cause loss of synaptic plasticity and induce neuronal cell death [62]. The ratio of Aβ 42/40 is a critical point of AD pathogenesis due to its more hydrophobic properties, causing it to be more prone to form oligomers and plaques [60].
Another hallmark in AD is NFT, which is caused by hyperphosphorylation of microtubule-associated protein tau (Mapt) inside the brain, which causes synaptic dysfunction and neuronal loss and leads to dementia [58,63]. Tau is a protein in human neurons that together with tubulin forms microtubules to stabilize the structure of the neuronal cell [64]. Its hyperphosphorylation forms a paired helix filament (PHF) in the brain [65]. On the other hand, hyperphosphorylated tau can bind to normal tau, MAP1, and MAP2 to induce deformation of micro-tubules, causing synaptic and axonal transport dysfunction [66]. Furthermore, insoluble NFT can alter the cytoplasmic function as well as axonal transport in the central nervous system, which leads to neuronal cell death and dementia progression [67].
Until now, AD was categorized as an incurable disease, but some treatments and medication can help to delay the progression [55]. The current treatment is to use cholinesterase inhibitors (ChEI) and partial N-methyl D-aspartate (NMDA) antagonist memantine, which are accepted by the Food and Drugs Administration (FDA) [68]. ChEI was first introduced in 1997 as a medication for mild and moderate AD. There are three types of drugs that are commonly used, namely, donepezil, galantamine, and rivastigmine [69]. ChEI inhibits cholinesterase, which cleaves the neurotransmitter acetylcholine (Ach) and terminates the function [70]. Memantine is an NMDA receptor antagonist that reduces the accumulation of calcium induced by NMDA receptor overstimulation in the neuronal cells [71]. Memantine is often used as a monotherapy or together with a low dose of acetylcholinesterase inhibitor (ACheI) in moderate and severe AD subjects [72]. A combination of memantine and donepezil significantly provides better outcomes on cognition and behavior improvement [73].

5. PD

PD is one of the known NDs, with its main features including loss of dopamine-producing neuronal cells in the substantia nigra and some others such as aggregation of α-synuclein protein in neurons and the presence of Lewy bodies in the brain [74,75]. It is characterized by motoric and non-motoric symptoms. The motoric symptoms include bradykinesia, resting tremor, rigidity, and postural instability as well as non-motoric symptoms such as hyposmia, sleep disturbances depression, orthostatic hypotension, constipation, and other dysautonomic symptoms [74,76]. Several risk factors such as age, gender, and ethnicity are associated with PD, but of all these risk factors, age is the greatest risk factor, wherein the prevalence and incidence of PD significantly increases with age [77]. The initiation factor is still unknown, but the progression of this disease is due to the loss of dopaminergic neuronal cells and some factors such as genetic, immune, and environmental factors [76,77]. Dopamine is one of the neurotransmitters that regulates several functions in the brain such as coordinated movement, emotion, and neuroendocrine secretion [78,79].
PD is mostly diagnosed by the presence of bradykinesia with resting tremor and/or rigidity [80]. Bradykinesia is a condition where the speed of spontaneous and repetitive movement is progressively reduced, and this condition usually happens in the early stage [81]. Before bradykinesia occurs in the patient, there is a condition called the prodromal stage, where nonmotor symptoms occur, such as constipation, loss of smell, sleep disorder, and several minor symptoms [82].
Medical therapies are the main treatment for PD, including pharmacotherapy and non-pharmacotherapy [83]. Dopamine receptor or intracerebral dopamine enhancer drugs are the main pharmacotherapies, such as levodopa, dopamine agonist, and monoamine oxidase-B inhibitor [75,83].

6. HD

HD is one type of ND that is inherited from parents to their offspring due to the increase of the CAG repeats huntingtin gene on chromosome 4 [84]. The risk of inheritance is equal for men and women, and the carrier’s gene develops the symptoms of this disease in normal life around the age of 40, but the onset of the disease might develop in childhood or teenage years [85,86]. There are several pathogenic mechanisms associated with HD due to the mutation of the Huntingtin protein and causing different brain damage [87]. HD is characterized by striatal degeneration in the brain, loss of medium spiny neurons, and atrophy in different regions of the brain, leading to distinct abnormal movements, psychiatric symptoms, and cognitive deficits that can be fatal 15–20 years after the disease onset [87,88,89].
Up to now, no medication can cure HD, but still, some drug and non-drug treatments can alleviate the symptoms of HD. Drug treatment is used to treat chorea, which is a movement disorder that causes unintended movement in HD patients [90]. Dopamine-reducing drugs such as tetrabenazine and/or antipsychotic agents such as risperidone and aripiprazole are usually used to treat chorea due to HD [84,90]. Non-drug treatment such as physiotherapy might be used to maintain the gait and balance of the subject for a longer period, and psychologists may also help to maintain the mental health of HD patients, which can help to reduce anxiety and depression [86,87].

7. Connection between H. pylori Infection and Neurodegenerative Diseases

There are several risk factors correlated with NDs, especially AD, such as age, traumatic head injury, depression, cardiovascular and cerebrovascular disease, and smoking. Recent studies showed that AD is also associated with H. pylori infection [91,92]. H. pylori is known to infect and cause several health problems in the human gastrointestinal (GI) tract such as gastric ulcers, gastritis, and gastric cancer [93]. In rare cases, a manifestation of extra gastric disease due to H. pylori infection might occur with several possible mechanism (Figure 3) and need to be taken into consideration. The extra gastric manifestation due to H. pylori infection, especially neurological problems, might occur through alteration of the gut–brain axis (GBA) [94]. The GBA is a bidirectional communication between the central nervous system (CNS) and enteric nervous system that integrates and links the gut and intestinal function with the central nervous system [95,96,97]. GBA modulates the GI function by regulating the GI immune system, mucosal change, and intestinal microbiome in response to stress and emotional and environmental influences [94,98].
H. pylori infection is associated with changes in gut microbiome composition [99]. Yang et al. demonstrated that children with gastritis showed an alteration of the gut microbiome, and this condition is worsened by the infection of H. pylori [100]. Zheng et al. also showed similar results, wherein in the H. pylori-positive subject, the abundance of Proteobacteria was increased while the abundances of other phyla such as Actinobacteria, Bacteroidetes, Firmicutes, Fusobacteria, Gemmatimonadetes, and Verrucomicrobia were significantly decreased compared to H. pylori-negative subject [101].
Alteration of the gut microbiome composition or so-called gut dysbiosis could lead to increased bacterial amyloid accumulation and intestinal innate immunity response, which induces systemic neuroinflammation, one of the hallmarks of AD [102]. The imbalance of the gut microbiome is related to increased gut permeability and gut barrier dysfunction, which causes toxic metabolites, bile acids, and pro-inflammatory cytokines to enter the circulatory system. The circulating toxic metabolites can reach the CNS and further cause leakage of the blood–brain barrier (BBB) and induce neuroinflammation due to microglia and astrocyte activation [103]. Doulberis et al. propose a hypothesis on how H. pylori might directly affect the CNS in three different ways: through the oral–nasal olfactory pathway, blood circulation by infecting monocytes and passing through the disrupted BBB, and the retrograde GI tract neural pathway [104].
Homocysteine (hcy) is one of the sulfur-containing amino acids that is derived from the demethylation process of methionine [105]. Hcy can be further processed into cysteine with the activity of cystathione-β-synthase enzyme and vitamin B6 as a cofactor. This reaction can occur when excess methionine is present in the body. In contrast, when the methionine level is low, hcy can be converted back to methionine by the remethylation process with the help of cofactor vitamin B6 and folic acid [106]. Hcy level in the human body usually ranges around 12–15 μmol/L, and elevation of hcy level is harmful to human bodies. This condition is known as hyperhomocysteinemia [105,107,108], elevated serum hcy is associated with neurological disorders such as cognitive decline, stroke, PD, and AD [109]. This condition can occur due to many factors such as lifestyle, administration of drugs and medication, or diseases such as chronic gastritis [110]. H. pylori infection is correlated with gastritis, and this condition can result in deficiency of vitamin B6 and folic acid. Deficiency of these vitamin cause the elevation of serum hcy level [111]. Elevated hcy levels can cause endothelial damage and result in atherothrombotic disorders and progression of AD [112].
Al-baret et al. showed that H. pylori infection in C57BL6 WT mice induced neuroinflammation by secretion of pro-inflammatory cytokines in the bloodstream without the deposition of amyloid plaques [113]. AD patients have a higher prevalence of H. pylori infection, and H. pylori antibodies are found in the cerebrospinal fluid (CSF) of AD patients [92,114]. Roubaud-Baudron et al. showed that H. pylori-infected AD subjects were more cognitively impaired and had higher neurodegenerative markers [115]. Wang et al. showed that H. pylori filtrate cultured with mouse neuroblastoma N2a cell and injected intraperitoneally into Sprague-Dawley rats induced AD-related tau hyper-phosphorylation in several sites such as Thr205, Thr231, and Ser404, together with the activation of glycogen synthase kinase-3β (GSK-3β) [116]. From the previous study, it might be concluded that H. pylori infection and AD might connected due to systemic inflammatory response and also through the gut–brain axis (GBA) interaction.
Apart from AD, PD and H. pylori might also correlate with each other through the GBA interaction. Changes in the gut microbiome might affect the metabolite production. As discussed, earlier H. pylori infection can induce the growth of Proteobacteria, which consists of mostly known pathogens [117]. Increased growth of pathogens will cause decreased production of short-chain fatty acids and increase the production of bacterial LPS [118]. LPS is the major constituent of the bacterial membrane in Gram-negative bacteria, which is an activator of inflammatory response [119]. LPS is predominantly recognized by Toll-like receptor (TLR) 4, which induces immune response and the release of pro-inflammatory cytokines [120]. H. pylori is known to express LPS, and it induces the production of cytokines, which may play a role in the pathogenesis of PD [1,118]. Altered gut microbiome composition also facilitates α-synuclein aggregate migration from the enteric nervous system (ENS) to the brain, causing progression of PD [121]. H. pylori infection also affects the absorption of drugs, especially levodopa, due to the change of intragastric pH [122,123].

8. Phytochemicals

Phytochemicals are non-nutritive bioactive compounds found in plants [14]. These bioactive compounds are the plant secondary metabolites that show health benefits for humans. Fruits, vegetables, grains, and nuts are the sources of natural phytochemicals. To understand the health benefits of these natural materials, these compounds need to be isolated and identified [124]. Different bioactive compounds show different mechanisms. The combined use of phytochemicals from different sources is needed to achieve greater health benefits. Phytochemicals are divided into several categories as phenolics, alkaloids, saponins, glucosinolates, terpenes, phytoestrogens, nitrogen-containing compounds, organosulfur compounds, carotenoids, and phytosterols [125,126].

8.1. Phenolics

Phenolics are one group of plant secondary metabolites consisting of at least one benzene ring and one hydroxyl group, playing important roles in benefitting health [127]. Phenolics can be divided into several subgroups up to the structures [125].
Fruits and vegetables are good sources of phenolics. Dark-colored fruits such as berries contain rich anthocyanins and flavonoids [128]. Cranberry contains 48 different polyphenols consisting of flavan-3-ols, flavonols, anthocyanins, phenolic acid, etc. [129]. Cranberry is rich in A-type proanthocyanidins, which provide health benefits [130]. Black raspberry contains high amounts of phenolics, mostly consisting of anthocyanins and ellagitannins [131]. The anthocyanin contents of black raspberries are the highest when compared with the other rubus species such as red raspberries and blackberry [132].

8.2. Carotenoids

There are hundreds of known carotenoids present in nature, but only a few of these carotenoids are good for humans [133]. Past studies showed that consumption of carotenoids was associated with a lower risk of eye problems, cancer, and cardiovascular diseases [134,135,136]. Carotenoids mostly consist of eight isoprenoid units with a total of 40 carbons as the backbone [137]. Carotenoids are divided into two major groups, carotenes (hydrocarbon carotenoids) and xanthophylls (oxygen-containing carotenoids) [138].
Past studies showed that high consumption of carotenoids, especially lycopene, can reduce the risk of cardiovascular diseases by decreasing low-density lipoprotein cholesterol and improving HDL function [139,140,141]. Another study also shows that the consumption of carotenoid-rich products could reduce visceral adiposity and ubiquinol (CoQ10) to prevent metabolic syndrome [142].

8.3. Alkaloids

Alkaloids are considered as all-nitrogen-containing compounds aside from peptides and their derivates, amines, cyanogenic glycosides, glucosinolates, cofactors, phytohormones, or primary metabolites [143]. Alkaloids can be classified according to different aspects such as biosynthesis pathways, chemical structure, and taxonomical groups [144].
According to previous studies, alkaloids exhibit different pharmacological activities, such as anti-microbial, anti-cancer, immunomodulatory, and antidiabetic effects [145,146,147,148,149,150].

8.4. Saponins

Saponins are bioactive compounds found in a wide variety of plants that are characterized by one or more sugar chains attached to steroid or triterpenoid aglycon backbone [151]. Saponins form foam when agitated in water due to their surface-active properties [152]. Saponins are synthesized from mevalonate primarily in the cytosol via farnesyl diphosphate and squalene [153]. Saponins have a low bioavailability due to their high molecular mass, hydrogen bonding capacity, and molecular flexibility [152].
Past studies have assessed different bioactivity of saponins. They can act as anti-bacterial and anti-fungal agents and act synergistically with antibiotics [154]. Marrelli et al. stated that saponins also exhibit antidiabetic activity by restoring insulin response and increasing insulin secretion from the pancreas [155].

9. Effect of Different Phytochemicals on H. pylori Infection

Natural phytochemicals in plants have been already assessed for their potency as anti-H. pylori substances (Figure 4). All the natural sources are discussed and summarized in vitro (Table 1) and in vivo (Table 2).
In vitro studies show that most of the phytochemicals reduce the inflammatory response by inhibiting the NF-κB activation and downregulating other pro-inflammatory cytokines [157,158,159]. Gingerol from ginger methanolic extract also inhibits the growth of 19 different strains of H. pylori, especially 5 CagA+ strains [156]. Furthermore, ginger shows anti-inflammatory effects and suppresses AP-1 activation [185]. Gaus et al. demonstrated that gingerol from ginger inhibited COX-2, transcription of NF-κB, and release of inflammatory cytokine release in a cell model and animal model [162].
Some other compounds also possess an anti-H. pylori effect to reduce the adhesion of H. pylori to epithelial cells in both in vitro and in vivo models. Polyphenols, especially flavonoids in monomer and oligomer, show a potential to reduce adhesion of H. pylori to epithelial cells [163,164,165,171]. Huang et al. demonstrated the removal of phenolics from noni fruit ethanolic and ethyl acetate extracts causing both extracts to lose antiadhesion activity of H. pylori to epithelial cells [143]. Polyphenols from apple peel extract also show the same effect in vitro and in vivo [142].
Cranberry showed anti-H. pylori activity due to its A-type proanthocyanidins content [186,187,188]. Gottenland et al. showed that cranberry extract together with Lactobacillus johnsonii La1 could reduce H. pylori infection. In vivo and in vitro studies show that cranberry can reduce the adhesion of H. pylori to epithelial cells and that A-type proanthocyanidin might play a critical role [24].
Essential oil derived from several herbs also can be used as an anti-H. pylori agent. Essential oil from Dittrichia viscosa shows potent anti-H. pylori activity with the major constituents being 3-methoxy cuminyl isobutyrate, α-cadinol, and α-eudesmol [160]. Another study by Ayoub et al., using Pimenta racemose essential oil that contains eugenol, was found to inhibit the growth of H. pylori together with inhibition of urease activity [177].

10. Effect of Different Phytochemicals on ND Development

NDs were associated with progressive neuronal cell death in the CNS [49]. Until now, there has been no medication to cure ND. Thus, only some medication can improve and delay the symptoms. Recent studies show that some phytochemicals would improve the condition of ND patients. Phytochemicals from natural sources have been assessed to show their potency as neuroprotective agents to improve ND (Figure 5). The neuroprotective activities of natural compounds are presented in Table 3 and Table 4.
Kim et al. and Park et al. showed that different types of curcuminoids, especially calebin-A, curcumin, demethoxycurcumin, bisdemethoxycurcumin, and 1,7-bis(4-hydroxyphenyl)-1-heptene-3,5-dione, can help to protect the PC12 cell line from Aβ insult in vitro, with the effective dose (ED50) ranging from 0.5 to 10.0 µg/mL [178,220]. Yang et al. demonstrated that curcumin can reduce the aggregation of Aβ and also disaggregate fibrillar Aβ40 fragments [221]. Ogunruku et al. found that polyphenol extracts from bell peppers can inhibit the activity of β-secretase (BACE 1) in a dose-dependent manner and also inhibit the aggregation of Aβ40 and reduce fibril formation in vitro [222]. Inflammation is the earliest sign of AD, which is induced by Aβ oligomer through many different receptors such as the Toll-like receptor (TLR) and formyl peptide receptor [223].
Animal studies also showed some phytochemical protection against AD. Purple berry rice contains rich anthocyanins and shows some improvement in AD by preventing memory impairment and hippocampal neurodegeneration in the Wistar rat model [202]. Anthocyanins from purpleberry rice reduces the activity of AChE, which can cleave the neurotransmitter acetylcholine and also reduce lipid peroxidation [70,202]. Another study using aqueous extract of white and red ginger also showed protection against AD in an animal model [203]. Both extracts can inhibit AChE activity and show a synergistic effect on inhibiting AChE activity. Furthermore, it also decreased sodium nitroprusside (SNP) and quinolinic acid (QA) elevated brain malondialdehyde (MDA) content, but there was no significant difference in SNP and QA lipid peroxidation in the brain. Ginsenosides found in ginseng could improve AD subject condition [214,215]. Both studies showed similar results on the improvement of the Alzheimer’s Diseases Assessment Scale (ADAS), Mini-Mental State Examination (MMSE), and Clinical Dementia Rating (CDR) scores in AD subjects. More importantly, results from Lee et al. also showed that after the discontinuation of ginseng powder from the treated group, scores of MMSE and ADAS were declining to the same as the control groups [182]. A high dose (990 mg/day) of cocoa flavonol for 8 weeks could improve the cognitive ability of elderly people with mild cognitive impairment [21].
Phytochemical treatment also provides improvement against other types of ND. Datla et al. demonstrated that treatment of tangeretin, a flavonoid derived from citrus fruit, can protect the neuronal cell from 6-hydroxydopamine (6-OHDA) toxicity in a rat model for PD [198]. Tangeretin can pass through the BBB and protect the dopaminergic neuronal cell, maintain TH+ cells, and significantly increase dopamine levels. Levites et al. demonstrated a positive result of green tea (−)-epigallocatechin-3-gallate (EGCG) treatment on N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson’s disease [197]. EGCG treatment protects the neuronal cell against MPTP toxicity, improves antioxidant enzyme activity, and significantly improves tyrosine hydroxylase (TH) activity. TH is an enzyme that plays a role in converting tyrosine into dopamine, and reduced TH activity can cause a reduction of dopamine synthesis and contribute to the progression of PD [224,225].
A previous study also shows improvement in HD by treatment of natural phytochemicals. Sandhir and Mehrotra demonstrated quercetin’s benefit for Huntington’s disease using an animal model, and their result showed that quercetin helps to reverse mitochondrial dysfunction due to 3-nitropropionic acid (3-NP) and reduces mitochondrial oxidative stress [205]. Mitochondrial function is considered to play an important role in the pathogenesis of neurodegenerative disease [226]. Dysfunction of mitochondria can cause alteration in the respiratory chain system, depletion of energy, and increased ROS production, which may lead to the progression of ND [227,228]. Kumar and Kumar also showed that two flavanones derived from citrus fruit, hesperidin and naringin, could protect the neuronal cell and improve mitochondrial function after treatment with 3-NP, and this effect further enhanced the protective effect of hesperidin and naringin when combined with L-NAME, which belong to NOS inhibitor [201].

11. H. pylori Eradication Improved Cognitive Function in an ND Subject

Until now, the association between H. pylori infection and ND is still controversial. Past studies showed that H. pylori infection has some common link with ND, especially after the eradication of H. pylori. Kountouras et al. showed that H. pylori eradication from AD patients showed some improvement in MMSE and Cambridge cognitive test scores compared to subjects who refused the H. pylori eradication therapy and H. pylori-negative subjects [229]. Furthermore, Chang et al. also showed that H. pylori eradication had a linkage with decreased progression of dementia in AD subjects [230]. H. pylori infection also interfere with the absorption of antiparkinsonian drugs, which could lead to worsened condition of PD [231]. The evidence obtained from previous studies might provide some research opportunities, especially for phytochemicals to eradicate H. pylori infection as well as improve cognitive function.

12. Conclusions

Studies showed that phytochemicals from natural sources would be used as anti-H. pylori and neuroprotective agents. These compounds can reduce the number of H. pylori and alleviate the inflammatory response due to H. pylori infection. Natural phytochemicals could be used as a therapeutical agent for H. pylori and neurodegenerative disease treatment due to their biological activity and safety concerns. Future studies are needed to find the potential and specific mechanism of each phytochemical in reducing H. pylori infection as well as the improvement of ND.

Author Contributions

C.-K.W. designed this review; Y.T., B.-K.C. and A.A. were involved in data collection; C.-K.W. and Y.T. drafted and formatted this review article; C.-K.W., Y.T., B.-K.C. and A.A. supervised and revised this review article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kusters, J.G.; van Vliet, A.H.M.; Kuipers, E.J. Pathogenesis of Helicobacter pylori infection. Clin. Microbiol. Rev. 2006, 19, 449–490. [Google Scholar] [CrossRef]
  2. Dunn, B.E.; Cohen, H.; Blaser, M.J.  Helicobacter pylori . Clin. Microbiol. Rev. 1997, 10, 720–741. [Google Scholar] [CrossRef] [PubMed]
  3. Kao, C.-Y.; Sheu, B.-S.; Wu, J.-J. Helicobacter pylori infection: An overview of bacterial virulence factors and pathogenesis. Biomed. J. 2016, 39, 14–23. [Google Scholar] [CrossRef] [PubMed]
  4. Selgrad, M.; Malfertheiner, P. Treatment of Helicobacter pylori. Curr. Opin. Gastroenterol. 2011, 27, 565–570. [Google Scholar] [CrossRef] [PubMed]
  5. Bytzer, P.; Dahlerup, J.F.; Eriksen, J.R.; Jarbøl, D.E.; Rosenstock, S.; Wildt, S.; Danish Society for Gastroenterology. Diagnosis and treatment of Helicobacter pylori infection. Dan. Med. Bull. 2011, 58, C4271. [Google Scholar] [PubMed]
  6. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
  7. Gitler, A.D.; Dhillon, P.; Shorter, J. Neurodegenerative disease: Models, mechanisms, and a new hope. Dis. Model Mech. 2017, 10, 499–502. [Google Scholar] [CrossRef]
  8. Chi, H.; Chang, H.-Y.; Sang, T.-K. Neuronal cell death mechanisms in major neurodegenerative diseases. Int. J. Mol. Sci. 2018, 19, 3082. [Google Scholar] [CrossRef]
  9. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  10. Mariani, E.; Polidori, M.C.; Cherubini, A.; Mecocci, P. Oxidative stress in brain aging, neurodegenerative and vascular diseases: An overview. J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 2005, 827, 65–75. [Google Scholar] [CrossRef]
  11. Beydoun, M.A.; Beydoun, H.A.; Elbejjani, M.; Dore, G.A.; Zonderman, A.B. Helicobacter pylori seropositivity and its association with incident all-cause and Alzheimer’s disease dementia in large national surveys. Alzheimer’s Dement. 2018, 14, 1148–1158. [Google Scholar] [CrossRef]
  12. Palacios, E.; Lobos-González, L.; Guerrero, S.; Kogan, M.J.; Shao, B.; Heinecke, J.W.; Quest, A.F.G.; Leyton, L.; Valenzuela-Valderrama, M. Helicobacter pylori outer membrane vesicles induce astrocyte reactivity through nuclear factor-κappa b activation and cause neuronal damage in vivo in a murine model. J. Neuroinflamm. 2023, 20, 66. [Google Scholar] [CrossRef] [PubMed]
  13. Mridula, K.R.; Borgohain, R.; Chandrasekhar Reddy, V.; Srinivasarao Bandaru, V.C.; Suryaprabha, T. Association of Helicobacter pylori with Parkinson’s disease. J. Clin. Neurol. 2017, 13, 181. [Google Scholar] [CrossRef]
  14. AL-Ishaq, R.K.; Overy, A.J.; Büsselberg, D. Phytochemicals and gastrointestinal cancer: Cellular mechanisms and effects to change cancer progression. Biomolecules 2020, 10, 105. [Google Scholar] [CrossRef] [PubMed]
  15. Shu, L.; Cheung, K.-L.; Khor, T.O.; Chen, C.; Kong, A.-N. Phytochemicals: Cancer chemoprevention and suppression of tumor onset and metastasis. Cancer Metastasis Rev. 2010, 29, 483–502. [Google Scholar] [CrossRef] [PubMed]
  16. Petrovska, B.B. Historical review of medicinal plants′ usage. Pharmacogn. Rev. 2012, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  17. Lima, M.C.; Paiva de Sousa, C.; Fernandez-Prada, C.; Harel, J.; Dubreuil, J.D.; de Souza, E.L. A review of the current evidence of fruit phenolic compounds as potential antimicrobials against pathogenic bacteria. Microb. Pathog. 2019, 130, 259–270. [Google Scholar] [CrossRef]
  18. Gregory, J.; Vengalasetti, Y.V.; Bredesen, D.E.; Rao, R.V. Neuroprotective herbs for the management of Alzheimer’s disease. Biomolecules 2021, 11, 543. [Google Scholar] [CrossRef]
  19. Önem, E.; Sarısu, H.C.; Özaydın, A.G.; Muhammed, M.T.; Ak, A. Phytochemical profile, antimicrobial, and anti-quorum sensing properties of fruit stalks of Prunus Avium L. Lett. Appl. Microbiol. 2021, 73, 426–437. [Google Scholar] [CrossRef]
  20. Li, Z.; Ma, J.; Guo, Y.; Liu, W.; Li, M.; Zhang, L.; Zhang, Y.; Zhou, T.; Zhang, J.; Gao, H.; et al. Suppression of Helicobacter pylori infection by daily cranberry intake: A double-blind, randomized, placebo-controlled trial. J. Gastroenterol. Hepatol. 2020, 36, 927–935. [Google Scholar] [CrossRef]
  21. Desideri, G.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Ghiadoni, L.; Mastroiacovo, D.; Raffaele, A.; Ferri, L.; Bocale, R.; Lechiara, M.; et al. Benefits in cognitive function, blood pressure, and insulin resistance through cocoa flavanol consumption in elderly subjects with mild cognitive impairment: The cocoa, cognition, and aging (cocoa) study. Hypertension 2012, 60, 794–801. [Google Scholar] [CrossRef] [PubMed]
  22. Kent, K.; Charlton, K.; Roodenrys, S.; Batterham, M.; Potter, J.; Traynor, V.; Gilbert, H.; Morgan, O.; Richards, R. Consumption of anthocyanin-rich cherry juice for 12 weeks improves memory and cognition in older adults with mild-to-moderate dementia. Eur. J. Nutr. 2015, 56, 333–341. [Google Scholar] [CrossRef] [PubMed]
  23. Marshall, B.J.; Armstrong, J.A.; McGechie, D.B.; Clancy, R.J. Attempt to fulfil Koch’s postulates for pyloric Campylobacter. Med. J. Aust. 1985, 142, 436–439. [Google Scholar] [CrossRef]
  24. Gotteland, M.; Andrews, M.; Toledo, M.; Muñoz, L.; Caceres, P.; Anziani, A.; Wittig, E.; Speisky, H.; Salazar, G. Modulation of Helicobacter pylori colonization with cranberry juice and Lactobacillus johnsonii La1 in children. Nutrition 2008, 24, 421–426. [Google Scholar] [CrossRef]
  25. Abbas, M.; Sharif, F.A.; Osman, S.M.; Osman, A.M.; El Sanousi, S.M.; Magzoub, M.; Ibrahim, M.E. Prevalence and associated symptoms of Helicobacter pylori infection among schoolchildren in Kassala state, east of Sudan. Interdiscip. Perspect. Infect. Dis. 2018, 2018, 4325752. [Google Scholar] [CrossRef] [PubMed]
  26. Palframan, S.L.; Kwok, T.; Gabriel, K. Vacuolating cytotoxin a (VacA), a key toxin for Helicobacter pylori pathogenesis. Front. Cell. Infect. Microbiol. 2012, 2, 92. [Google Scholar] [CrossRef]
  27. Abdullah, M.; Greenfield, L.K.; Bronte-Tinkew, D.; Capurro, M.I.; Rizzuti, D.; Jones, N.L. VacA promotes CagA accumulation in gastric epithelial cells during Helicobacter pylori Infection. Sci. Rep. 2019, 9, 38. [Google Scholar] [CrossRef]
  28. Willhite, D.C.; Cover, T.L.; Blanke, S.R. Cellular vacuolation and mitochondrial cytochrome c release are independent outcomes of Helicobacter pylori vacuolating cytotoxin activity that are each dependent on membrane channel formation. J. Biol. Chem. 2003, 278, 48204–48209. [Google Scholar] [CrossRef]
  29. Rao, R.V.; Peel, A.; Logvinova, A.; del Rio, G.; Hermel, E.; Yokota, T.; Goldsmith, P.C.; Ellerby, L.M.; Ellerby, H.M.; Bredesen, D.E. Coupling endoplasmic reticulum stress to the cell death program: Role of the er chaperone grp78. FEBS Lett. 2002, 514, 122–128. [Google Scholar] [CrossRef]
  30. Fulda, S.; Debatin, K.-M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef]
  31. Rassow, J. Helicobacter pylori vacuolating toxin A and apoptosis. Cell Commun. Signal. 2011, 9, 26. [Google Scholar] [CrossRef]
  32. Foegeding, N.J.; Caston, R.R.; McClain, M.S.; Ohi, M.D.; Cover, T.L. An overview of Helicobacter pylori VacA toxin biology. Toxins 2016, 8, 173. [Google Scholar] [CrossRef]
  33. Jiménez-Soto, L.F.; Haas, R. The CagA toxin of Helicobacter pylori: Abundant production but relatively low amount translocated. Sci. Rep. 2016, 6, 23227. [Google Scholar] [CrossRef]
  34. Hatakeyama, M. Structure and function of Helicobacter pylori CagA, the first-identified bacterial protein involved in human cancer. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2017, 93, 196–219. [Google Scholar] [CrossRef]
  35. Jones, K.R.; Whitmire, J.M.; Merrell, D.S. A Tale of two toxins: Helicobacter pylori CagA and VacA modulate host pathways that impact disease. Front. Microbiol. 2010, 1, 23227. [Google Scholar] [CrossRef]
  36. Suzuki, M.; Mimuro, H.; Kiga, K.; Fukumatsu, M.; Ishijima, N.; Morikawa, H.; Nagai, S.; Koyasu, S.; Gilman, R.H.; Kersulyte, D.; et al. Helicobacter pylori CagA phosphorylation-independent function in epithelial proliferation and inflammation. Cell Host Microbe 2009, 5, 23–34. [Google Scholar] [CrossRef] [PubMed]
  37. Mobley, H.; The role of Helicobacter pylori urease in the pathogenesis of gastritis and peptic ulceration. Aliment. Pharmacol. Ther. 1996, 10 (Suppl. S1), 57–64. [Google Scholar]
  38. Olivera-Severo, D.; Uberti, A.F.; Marques, M.S.; Pinto, M.T.; Gomez-Lazaro, M.; Figueiredo, C.; Leite, M.; Carlini, C.R. A New Role for Helicobacter pylori urease: Contributions to angiogenesis. Front. Microbiol. 2017, 8, 1883. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, W.; Xu, L.; Xu, C. Relationship between Helicobacter pylori infection and gastrointestinal microecology. Front. Cell. Infect. Microbiol. 2022, 12, 938608. [Google Scholar] [CrossRef] [PubMed]
  40. Mišak, Z.; Hojsak, I.; Homan, M. Review: Helicobacter pylori in pediatrics. Helicobacter 2019, 24, e12639. [Google Scholar] [CrossRef] [PubMed]
  41. Araújo, G.R.L.; Marques, H.S.; Santos, M.L.C.; da Silva, F.A.F.; de Brito, B.B.; Santos, G.L.C.; de Melo, F.F. Helicobacter pylori infection: How does age influence the inflammatory pattern? World J. Gastroenterol. 2022, 28, 402–411. [Google Scholar] [CrossRef] [PubMed]
  42. Meliț, L.E.; Mărginean, C.O.; Mărginean, C.D.; Mărginean, M.O. The relationship between toll-like receptors and Helicobacter pylori-related gastropathies: Still a controversial topic. J. Immunol. Res. 2019, 2019, 8197048. [Google Scholar] [CrossRef]
  43. Kalali, B.; Mejías-Luque, R.; Javaheri, A.; Gerhard, M. H. pylori Virulence factors: Influence on immune system and pathology. Mediators Inflamm. 2014, 2014, 426309. [Google Scholar] [CrossRef] [PubMed]
  44. Logan, R.P. Urea Breath Tests in the Management of Helicobacter pylori infection. Gut 1998, 43 (Suppl. S1), S47–S50. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, H.; Hu, B. Diagnosis of Helicobacter pylori infection and recent advances. Diagnostics 2021, 11, 1305. [Google Scholar] [CrossRef]
  46. Graham, D.Y.; Miftahussurur, M. Helicobacter pylori urease for diagnosis of Helicobacter pylori infection: A mini review. J. Adv. Res. 2018, 13, 51–57. [Google Scholar] [CrossRef] [PubMed]
  47. Blumenthal, K.G.; Peter, J.G.; Trubiano, J.A.; Phillips, E.J. Antibiotic allergy. Lancet 2019, 393, 183–198. [Google Scholar] [CrossRef]
  48. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022, 11, e1260. [Google Scholar] [CrossRef]
  49. Kovacs, G.G. Concepts and classification of neurodegenerative diseases. Handb. Clin. Neurol. 2018, 145, 301–307. [Google Scholar] [CrossRef]
  50. Rachakonda, V.; Pan, T.H.; Le, W.D. Biomarkers of neurodegenerative disorders: How good are they? Cell Res. 2004, 14, 349–358. [Google Scholar] [CrossRef]
  51. Duncan, G.W. The aging brain and neurodegenerative diseases. Clin. Geriatr. Med. 2011, 27, 629–644. [Google Scholar] [CrossRef]
  52. Pierre, G. Neurodegenerative disorders and metabolic disease. Arch. Dis. Child. 2013, 98, 618–624. [Google Scholar] [CrossRef] [PubMed]
  53. Kawamata, H.; Manfredi, G. Introduction to neurodegenerative diseases and related techniques. Methods Mol. Biol. 2011, 793, 3–8. [Google Scholar] [CrossRef] [PubMed]
  54. Manoharan, S.; Guillemin, G.J.; Abiramasundari, R.S.; Essa, M.M.; Akbar, M.; Akbar, M.D. The role of reactive oxygen species in the pathogenesis of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease: A mini review. Oxid. Med. Cell. Longev. 2016, 2016, 8590578. [Google Scholar] [CrossRef]
  55. Kumar, A.; Sidhu, J.; Goyal, A.; Tsao, J.W. Alzheimer Disease. In Statpearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://pubmed.ncbi.nlm.nih.gov/29763097/ (accessed on 22 September 2022).
  56. Budelier, M.M.; Bateman, R.J. Biomarkers of Alzheimer disease. J. Appl. Lab. Med. 2019, 5, 194–208. [Google Scholar] [CrossRef] [PubMed]
  57. Soria Lopez, J.A.; González, H.M.; Léger, G.C. Alzheimer’s disease. Handb. Clin. Neurol. 2019, 167, 231–255. [Google Scholar] [CrossRef]
  58. Kametani, F.; Hasegawa, M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front. Neurosci. 2018, 12, 25. [Google Scholar] [CrossRef]
  59. Rukmangadachar, L.A.; Bollu, P.C. Amyloid beta peptide. In Statpearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://pubmed.ncbi.nlm.nih.gov/29083757/ (accessed on 12 October 2022).
  60. Zhang, Y.; Thompson, R.; Zhang, H.; Xu, H. App processing in Alzheimer’s disease. Mol. Brain 2011, 4, 3. [Google Scholar] [CrossRef]
  61. Cras, P.; Kawai, M.; Lowery, D.; Gonzalez-DeWhitt, P.; Greenberg, B.; Perry, G. Senile Plaque Neurites in Alzheimer Disease Accumulate Amyloid Precursor Protein. Proc. Natl. Acad. Sci. USA 1991, 88, 7552–7556. [Google Scholar] [CrossRef]
  62. Khan, S.; Barve, K.H.; Kumar, M.S. Recent advancements in pathogenesis, diagnostics and treatment of Alzheimer’s disease. Curr. Neuropharmacol. 2020, 18, 1106–1125. [Google Scholar] [CrossRef]
  63. Kempf, S.J.; Metaxas, A. Neurofibrillary tangles in Alzheimer’s disease: Elucidation of the molecular mechanism by immunohistochemistry and tau protein phospho-proteomics. Neural Regen. Res. 2016, 11, 1579. [Google Scholar] [CrossRef]
  64. Iqbal, K.; Liu, F.; Gong, C.-X.; Grundke-Iqbal, I. Tau in Alzheimer disease and related tauopathies. Curr. Alzheimer Res. 2010, 7, 656–664. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, J.-Z.; Xia, Y.-Y.; Grundke-Iqbal, I.; Iqbal, K. Abnormal hyperphosphorylation of tau: Sites, regulation, and molecular mechanism of neurofibrillary degeneration. J. Alzheimer’s Dis. 2013, 33 (Suppl. S1), S123–S139. [Google Scholar] [CrossRef]
  66. Iqbal, K.; Gong, C.-X.; Liu, F. Hyperphosphorylation-induced tau oligomers. Front. Neurol. 2013, 4, 112. [Google Scholar] [CrossRef]
  67. Mudher, A.; Lovestone, S. Alzheimer’s disease—Do Taoists and Baptists finally shake hands? Trends Neurosci. 2002, 25, 22–26. [Google Scholar] [CrossRef] [PubMed]
  68. Vaz, M.; Silvestre, S. Alzheimer’s disease: Recent treatment strategies. Eur. J. Pharmacol. 2020, 887, 173554. [Google Scholar] [CrossRef] [PubMed]
  69. Birks, J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst. Rev. 2006, 1, CD005593. [Google Scholar] [CrossRef]
  70. Pohanka, M. Cholinesterases, a target of pharmacology and toxicology. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc 2011, 155, 219–223. [Google Scholar] [CrossRef]
  71. Robinson, D.M.; Keating, G.M. Memantine: A review of its use in Alzheimer’s disease. Drugs 2006, 66, 1515–1534. [Google Scholar] [CrossRef]
  72. McKeage, K. Memantine. CNS Drugs 2009, 23, 881–897. [Google Scholar] [CrossRef]
  73. Tariot, P.N.; Farlow, M.R.; Grossberg, G.T.; Graham, S.M.; McDonald, S.; Gergel, I.; for the Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil. JAMA 2004, 291, 317. [Google Scholar] [CrossRef]
  74. Lew, M. Overview of Parkinson’s disease. Pharmacotherapy 2007, 27 Pt 2, 155S160S. [Google Scholar] [CrossRef]
  75. Armstrong, M.J.; Okun, M.S. Diagnosis and treatment of Parkinson disease. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef] [PubMed]
  76. Antony, P.M.A.; Diederich, N.J.; Krüger, R.; Balling, R. The hallmarks of Parkinson’s disease. FEBS J. 2013, 280, 5981–5993. [Google Scholar] [CrossRef] [PubMed]
  77. De Virgilio, A.; Greco, A.; Fabbrini, G.; Inghilleri, M.; Rizzo, M.I.; Gallo, A.; Conte, M.; Rosato, C.; Ciniglio Appiani, M.; de Vincentiis, M. Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun. Rev. 2016, 15, 1005–1011. [Google Scholar] [CrossRef] [PubMed]
  78. Jaber, M.; Robinson, S.W.; Missale, C.; Caron, M.G. Dopamine receptors and brain function. Neuropharmacology 1996, 35, 1503–1519. [Google Scholar] [CrossRef] [PubMed]
  79. Basu, S.; Dasgupta, P.S. Dopamine, a neurotransmitter, influences the immune system. J. Neuroimmunol. 2000, 102, 113–124. [Google Scholar] [CrossRef]
  80. Reich, S.G.; Savitt, J.M. Parkinson’s disease. Med. Clin. N. Am. 2019, 103, 337–350. [Google Scholar] [CrossRef]
  81. Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
  82. Tolosa, E.; Garrido, A.; Scholz, S.W.; Poewe, W. Challenges in the diagnosis of Parkinson’s disease. Lancet Neurol. 2021, 20, 385–397. [Google Scholar] [CrossRef]
  83. Beitz, J.M. Parkinson’s disease: A review. Front Biosci Schol Ed. 2014, 6, 65–74. [Google Scholar] [CrossRef]
  84. Snowden, J.S. The neuropsychology of Huntington’s disease. Arch. Clin. Neuropsychol. 2017, 32, 876–887. [Google Scholar] [CrossRef]
  85. Paulsen, J.S.; Langbehn, D.R.; Stout, J.C.; Aylward, E.; Ross, C.A.; Nance, M.; Guttman, M.; Johnson, S.; MacDonald, M.; Beglinger, L.J.; et al. Detection of Huntington’s disease decades before diagnosis: The predict-HD study. J. Neurol. Neurosurg. Psychiatry 2008, 79, 874–880. [Google Scholar] [CrossRef]
  86. Read, J.; Jones, R.; Owen, G.; Leavitt, B.R.; Coleman, A.; Roos, R.A.C.; Dumas, E.M.; Durr, A.; Justo, D.; Say, M.; et al. Quality of life in Huntington’s disease: A comparative study investigating the impact for those with pre-manifest and early manifest disease, and their partners. J. Huntington’s Dis. 2013, 2, 159–175. [Google Scholar] [CrossRef]
  87. Ghosh, R.; Tabrizi, S.J. Clinical features of Huntington’s disease. Adv. Exp. Med. Biol. 2018, 1049, 1–28. [Google Scholar] [CrossRef]
  88. McColgan, P.; Tabrizi, S.J. Huntington’s disease: A clinical review. Eur. J. Neurol. 2017, 25, 24–34. [Google Scholar] [CrossRef] [PubMed]
  89. Pan, L.; Feigin, A. Huntington’s disease: New frontiers in therapeutics. Curr. Neurol. Neurosci. Rep. 2021, 21, 10. [Google Scholar] [CrossRef]
  90. Stoker, T.B.; Mason, S.L.; Greenland, J.C.; Holden, S.T.; Santini, H.; Barker, R.A. Huntington’s disease: Diagnosis and management. Pract. Neurol. 2022, 22, 32–41. [Google Scholar] [CrossRef] [PubMed]
  91. Doulberis, M.; Saleh, C.; Beyenburg, S. Is There an association between migraine and gastrointestinal disorders? J. Clin. Neurol. 2017, 13, 215. [Google Scholar] [CrossRef]
  92. Kountouras, J.M.D.P.; Tsolaki, M.; Gavalas, E.; Boziki, M.; Zavos, C.; Karatzoglou, P.; Chatzopoulos, D.; Venizelos, I. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology 2006, 66, 938–940. [Google Scholar] [CrossRef]
  93. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global prevalence of Helicobacter pylori infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef] [PubMed]
  94. Budzyński, J.; Kłopocka, M. Brain-gut axis in the pathogenesis of Helicobacter pylori infection. World J. Gastroenterol. 2014, 20, 5212. [Google Scholar] [CrossRef]
  95. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  96. Zhou, L.; Foster, J.A. Psychobiotics and the gut–brain axis: In the pursuit of happiness. Neuropsychiatr. Dis. Treat. 2015, 11, 715. [Google Scholar] [CrossRef]
  97. Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The gut microbiota–brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2020, 19, 241–255. [Google Scholar] [CrossRef]
  98. Megur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The microbiota–gut–brain axis and Alzheimer’s disease: Neuroinflammation is to blame? Nutrients 2020, 13, 37. [Google Scholar] [CrossRef]
  99. Iino, C.; Shimoyama, T. Impact of Helicobacter pylori infection on gut microbiota. World J. Gastroenterol. 2021, 27, 6224–6230. [Google Scholar] [CrossRef]
  100. Yang, L.; Zhang, J.; Xu, J.; Wei, X.; Yang, J.; Liu, Y.; Li, H.; Zhao, C.; Wang, Y.; Zhang, L.; et al. Helicobacter pylori infection aggravates dysbiosis of gut microbiome in children with gastritis. Front. Cell. Infect. Microbiol. 2019, 9, 375. [Google Scholar] [CrossRef]
  101. Zheng, W.; Miao, J.; Luo, L.; Long, G.; Chen, B.; Shu, X.; Gu, W.; Peng, K.; Li, F.; Zhao, H.; et al. The effects of Helicobacter pylori infection on microbiota associated with gastric mucosa and immune factors in children. Front. Immunol. 2021, 12, 625586. [Google Scholar] [CrossRef]
  102. Shen, L.; Liu, L.; Ji, H.-F. Alzheimer’s disease histological and behavioral manifestations in transgenic mice correlate with specific gut microbiome state. J. Alzheimer’s Dis. 2017, 56, 385–390. [Google Scholar] [CrossRef]
  103. Zou, B.; Li, J.; Ma, R.-X.; Cheng, X.-Y.; Ma, R.-Y.; Zhou, T.-Y.; Wu, Z.-Q.; Yao, Y.; Li, J. Gut microbiota is an impact factor based on the brain-gut axis to alzheimer’s disease: A systematic review. Aging Dis. 2023, 14, 964. [Google Scholar] [CrossRef]
  104. Doulberis, M.; Kotronis, G.; Thomann, R.; Polyzos, S.A.; Boziki, M.; Gialamprinou, D.; Deretzi, G.; Katsinelos, P.; Kountouras, J. Review: Impact of Helicobacter pylori on Alzheimer’s disease: What do we know so far? Helicobacter 2017, 23, e12454. [Google Scholar] [CrossRef]
  105. Tawfik, A.; Samra, Y.A.; Elsherbiny, N.M.; Al-Shabrawey, M. Implication of hyperhomocysteinemia in blood retinal barrier (BRB) dysfunction. Biomolecules 2020, 10, E1119. [Google Scholar] [CrossRef]
  106. Brustolin, S.; Giugliani, R.; Félix, T.M. Genetics of homocysteine metabolism and associated disorders. Braz. J. Med. Biol. Res. 2010, 43, 1–7. [Google Scholar] [CrossRef] [PubMed]
  107. Kumar, A.; Palfrey, H.A.; Pathak, R.; Kadowitz, P.J.; Gettys, T.W.; Murthy, S.N. The metabolism and significance of homocysteine in nutrition and health. Nutr. Metab. 2017, 14, 78. [Google Scholar] [CrossRef] [PubMed]
  108. Selhub, J. Homocysteine metabolism. Annu. Rev. Nutr. 1999, 19, 217–246. [Google Scholar] [CrossRef] [PubMed]
  109. Tinelli, C.; Di Pino, A.; Ficulle, E.; Marcelli, S.; Feligioni, M. Hyperhomocysteinemia as a risk factor and potential nutraceutical target for certain pathologies. Front. Nutr. 2019, 6, 49. [Google Scholar] [CrossRef]
  110. Smith, A.D.; Refsum, H.; Bottiglieri, T.; Fenech, M.; Hooshmand, B.; McCaddon, A.; Miller, J.W.; Rosenberg, I.H.; Obeid, R. Homocysteine and dementia: An international consensus statement. J. Alzheimer’s Dis. 2018, 62, 561–570. [Google Scholar] [CrossRef] [PubMed]
  111. Kountouras, J.; Gavalas, E.; Boziki, M.; Zavos, C. Helicobacter pylori may be involved in cognitive impairment and dementia development through induction of atrophic gastritis, vitamin b-12–folate deficiency, and hyperhomocysteinemia sequence. Am. J. Clin. Nutr. 2007, 86, 805–806. [Google Scholar] [CrossRef]
  112. Kountouras, J.; Gavalas, E.; Zavos, C.; Stergiopoulos, C.; Chatzopoulos, D.; Kapetanakis, N.; Gisakis, D. Alzheimer’s disease and Helicobacter pylori infection: Defective immune regulation and apoptosis as proposed common links. Med. Hypotheses 2007, 68, 378–388. [Google Scholar] [CrossRef]
  113. Albaret, G.; Sifré, E.; Floch, P.; Laye, S.; Aubert, A.; Dubus, P.; Azzi-Martin, L.; Giese, A.; Salles, N.; Mégraud, F.; et al. Alzheimer’s disease and Helicobacter pylori infection: Inflammation from stomach to brain? J. Alzheimer’s Dis. 2020, 73, 801–809. [Google Scholar] [CrossRef]
  114. Malaguarnera, M.; Bella, R.; Alagona, G.; Ferri, R.; Carnemolla, A.; Pennisi, G. Helicobacter pylori and Alzheimer’s disease: A possible link. Eur. J. Intern. Med. 2004, 15, 381–386. [Google Scholar] [CrossRef]
  115. Roubaud-Baudron, C.; Krolak-Salmon, P.; Quadrio, I.; Mégraud, F.; Salles, N. Impact of chronic Helicobacter pylori infection on Alzheimer’s disease: Preliminary results. Neurobiol. Aging 2012, 33, 1009.e11–1009.e19. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, X.-L.; Zeng, J.; Yang, Y.; Xiong, Y.; Zhang, Z.-H.; Qiu, M.; Yan, X.; Sun, X.-Y.; Tuo, Q.-Z.; Liu, R.; et al. Helicobacter pylori filtrate induces Alzheimer-like tau hyperphosphorylation by activating glycogen synthase kinase-3β. J. Alzheimer’s Dis. 2014, 43, 153–165. [Google Scholar] [CrossRef]
  117. Rizzatti, G.; Lopetuso, L.R.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A common factor in human diseases. Biomed Res. Int. 2017, 2017, 9351507. [Google Scholar] [CrossRef]
  118. Toledo, A.R.L.; Monroy, G.R.; Salazar, F.E.; Lee, J.-Y.; Jain, S.; Yadav, H.; Borlongan, C.V. Gut–brain axis as a pathological and therapeutic target for neurodegenerative disorders. Int. J. Mol. Sci. 2022, 23, 1184. [Google Scholar] [CrossRef]
  119. Rhee, S.H. Lipopolysaccharide: Basic biochemistry, intracellular signaling, and physiological impacts in the gut. Intest. Res. 2014, 12, 90–95. [Google Scholar] [CrossRef] [PubMed]
  120. Zielen, S.; Trischler, J.; Schubert, R. Lipopolysaccharide challenge: Immunological effects and safety in humans. Expert Rev. Clin. Immunol. 2015, 11, 409–418. [Google Scholar] [CrossRef] [PubMed]
  121. Lubomski, M.; Tan, A.H.; Lim, S.-Y.; Holmes, A.J.; Davis, R.L.; Sue, C.M. Parkinson’s disease and the gastrointestinal microbiome. J. Neurol. 2019, 267, 2507–2523. [Google Scholar] [CrossRef]
  122. Çamcı, G.; Oğuz, S. Association between Parkinson’s disease and Helicobacter pylori. J. Clin. Neurol. 2016, 12, 147. [Google Scholar] [CrossRef]
  123. Lahner, E.; Annibale, B.; Delle Fave, G. Systematic review: Heliocobacter pylori infection and impaired drug absorption. Aliment. Pharmacol. Ther. 2009, 29, 379–386. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, R.H. Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. Am. J. Clin. Nutr. 2003, 78, 517S–520S. [Google Scholar] [CrossRef]
  125. Liu, R.H. Health-promoting components of fruits and vegetables in the diet. Adv. Nutr. 2013, 4, 384S–392S. [Google Scholar] [CrossRef]
  126. Leitzmann, C. Characteristics and health benefits of phytochemicals. Forsch. Komplementmed. 2016, 23, 69–74. [Google Scholar] [CrossRef] [PubMed]
  127. Van Hung, P. Phenolic compounds of cereals and their antioxidant capacity. Crit. Rev. Food Sci. Nutr. 2014, 56, 25–35. [Google Scholar] [CrossRef] [PubMed]
  128. Skrovankova, S.; Sumczynski, D.; Mlcek, J.; Jurikova, T.; Sochor, J. Bioactive compounds and antioxidant activity in different types of berries. Int. J. Mol. Sci. 2015, 16, 24673–24706. [Google Scholar] [CrossRef] [PubMed]
  129. Oszmiański, J.; Lachowicz, S.; Józef Gorzelany; Matłok, N. The effect of different maturity stages on phytochemical composition and antioxidant capacity of cranberry cultivars. Eur. Food Res. Technol. 2017, 244, 705–719. [Google Scholar] [CrossRef]
  130. Oszmiański, J.; Kolniak-Ostek, J.; Lachowicz, S.; Gorzelany, J.; Matłok, N. Phytochemical compounds and antioxidant activity in different cultivars of cranberry (Vaccinium macrocarpon L). J. Food Sci. 2017, 82, 2569–2575. [Google Scholar] [CrossRef]
  131. Kula, M.; Krauze-Baranowska, M. Rubus occidentalis: The black raspberry—Its potential in the prevention of cancer. Nutr. Cancer 2015, 68, 18–28. [Google Scholar] [CrossRef] [PubMed]
  132. Tian, Q.; Giusti, M.M.; Stoner, G.D.; Schwartz, S.J. Characterization of a new anthocyanin in black raspberries (Rubus occidentalis) by liquid chromatography electrospray ionization tandem mass spectrometry. Food Chem. 2006, 94, 465–468. [Google Scholar] [CrossRef]
  133. Tanumihardjo, S.A. Carotenoids: Health effects. In Encyclopedia of Human Nutrition; Elsevier: Amsterdam, The Netherlands, 2013; Available online: https://www.sciencedirect.com/science/article/abs/pii/B9780123750839000453?via%3Dihub (accessed on 26 October 2022).
  134. Miller, E.C.; Giovannucci, E.; Erdman, J.W.; Bahnson, R.; Schwartz, S.J.; Clinton, S.K. Tomato products, lycopene, and prostate cancer risk. Urol. Clin. North Am. 2002, 29, 83–93. [Google Scholar] [CrossRef]
  135. Voutilainen, S.; Nurmi, T.; Mursu, J.; Rissanen, T.H. Carotenoids and cardiovascular health. Am. J. Clin. Nutr. 2006, 83, 1265–1271. [Google Scholar] [CrossRef]
  136. Abdel-Aal, E.-S.M.; Akhtar, H.; Zaheer, K.; Ali, R. Dietary sources of lutein and zeaxanthin carotenoids and their role in eye health. Nutrients 2013, 5, 1169–1185. [Google Scholar] [CrossRef] [PubMed]
  137. Rodriguez-Amaya, D.B. Structures and analysis of carotenoid molecules. Subcell. Biochem. 2016, 79, 71–108. [Google Scholar] [CrossRef] [PubMed]
  138. Misawa, N. 1.20—Carotenoids. In Comprehensive Natural Products II; Elsevier: Amsterdam, The Netherlands, 2010; Available online: https://www.sciencedirect.com/science/article/abs/pii/B9780080453828000095?via%3Dihub (accessed on 20 February 2023).
  139. Visioli, F.; Riso, P.; Grande, S.; Galli, C.; Porrini, M. Protective activity of tomato products on in vivo markers of lipid oxidation. Eur. J. Nutr. 2003, 42, 201–206. [Google Scholar] [CrossRef] [PubMed]
  140. Palozza, P.; Catalano, A.; Simone, R.E.; Mele, M.C.; Cittadini, A. Effect of lycopene and tomato products on cholesterol metabolism. Ann. Nutr. Metab. 2012, 61, 126–134. [Google Scholar] [CrossRef]
  141. McEneny, J.; Wade, L.; Young, I.S.; Masson, L.; Duthie, G.; McGinty, A.; McMaster, C.; Thies, F. Lycopene intervention reduces inflammation and improves HDL functionality in moderately overweight middle-aged individuals. J. Nutr. Biochem. 2013, 24, 163–168. [Google Scholar] [CrossRef]
  142. Takagi, T.; Hayashi, R.; Nakai, Y.; Okada, S.; Miyashita, R.; Yamada, M.; Mihara, Y.; Mizushima, K.; Morita, M.; Uchiyama, K.; et al. Dietary intake of carotenoid-rich vegetables reduces visceral adiposity in obese Japanese men—A randomized, double-blind trial. Nutrients 2020, 12, 2342. [Google Scholar] [CrossRef]
  143. Wink, M. Alkaloids: Properties and Determination. In Encyclopedia of Food and Health; Elsevier: Amsterdam, The Netherlands, 2010; Available online: https://www.sciencedirect.com/science/article/abs/pii/B9780123849472000192 (accessed on 11 October 2023).
  144. Bhambhani, S.; Kondhare, K.R.; Giri, A.P. Diversity in chemical structures and biological properties of plant alkaloids. Molecules 2021, 26, 3374. [Google Scholar] [CrossRef]
  145. Makarieva, T.N.; Dmitrenok, A.S.; Dmitrenok, P.S.; Grebnev, B.B.; Stonik, V.A. Pibocin B, the first N-O-methylindole marine alkaloid, a metabolite from the far-eastern ascidian Eudistoma species. J. Nat. Prod. 2001, 64, 1559–1561. [Google Scholar] [CrossRef]
  146. Chay, C.; Cansino, R.; Pinzón, C.; Torres-Ochoa, R.; Martínez, R. Synthesis and anti-tuberculosis activity of the marine natural product caulerpin and its analogues. Mar. Drugs 2014, 12, 1757–1772. [Google Scholar] [CrossRef]
  147. Reyes, F.; Fernández, R.; Rodríguez, A.; Bueno, S.; de Eguilior, C.; Francesch, A.; Cuevas, C. Cytotoxic staurosporines from the marine ascidian Cystodytes solitus. J. Nat. Prod. 2008, 71, 1046–1048. [Google Scholar] [CrossRef]
  148. Gul, W.; Hamann, M.T. Indole alkaloid marine natural products: An established source of cancer drug leads with considerable promise for the control of parasitic, neurological and other diseases. Life Sci. 2005, 78, 442–453. [Google Scholar] [CrossRef] [PubMed]
  149. Lind, K.F.; Østerud, B.; Hansen, E.; Jørgensen, T.Ø.; Andersen, J.H. The immunomodulatory effects of barettin and involvement of the kinases CAMK1α and RIPK2. Immunopharmacol. Immunotoxicol. 2015, 37, 458–464. [Google Scholar] [CrossRef] [PubMed]
  150. Mao, S.-C.; Guo, Y.-W.; Shen, X. Two novel aromatic valerenane-type sesquiterpenes from the chinese green alga Caulerpa taxifolia. Bioorg. Med. Chem. Lett. 2006, 16, 2947–2950. [Google Scholar] [CrossRef] [PubMed]
  151. Güçlü-Ustündağ, O.; Mazza, G. Saponins: Properties, applications and processing. Crit. Rev. Food Sci. Nutr. 2007, 47, 231–258. [Google Scholar] [CrossRef] [PubMed]
  152. Yakindra Prasad Timilsena; Arissara Phosanam; Stockmann, R. Perspectives on saponins: Food functionality and applications. Int. J. Mol. Sci. 2023, 24, 13538. [Google Scholar] [CrossRef]
  153. Osbourn, A.; Goss, R.J.M.; Field, R.A. The saponins: Polar isoprenoids with important and diverse biological activities. Nat. Prod. Rep. 2011, 28, 1261–1268. [Google Scholar] [CrossRef]
  154. Tagousop, C.N.; Tamokou, J.-D.; Kengne, I.C.; Ngnokam, D.; Voutquenne-Nazabadioko, L. Antimicrobial activities of saponins from Melanthera elliptica and their synergistic effects with antibiotics against pathogenic phenotypes. Chem. Cent. J. 2018, 12, 97. [Google Scholar] [CrossRef] [PubMed]
  155. Marrelli, M.; Conforti, F.; Araniti, F.; Statti, G. Effects of saponins on lipid metabolism: A review of potential health benefits in the treatment of obesity. Molecules 2016, 21, 1404. [Google Scholar] [CrossRef]
  156. Mahady, G.B.; Pendland, S.L.; Yun, G.S.; Lu, Z.-Z.; Stoia, A. Ginger (Zingiber officinale Roscoe) and the gingerols inhibit the growth of CagA+ strains of Helicobacter pylori. Anticancer Res. 2003, 23, 3699–3702. [Google Scholar]
  157. Foryst-Ludwig, A.; Neumann, M.; Schneider-Brachert, W.; Naumann, M. Curcumin blocks NF-κB and the motogenic response in Helicobacter pylori infected epithelial cells. Biochem. Biophys. Res. Commun. 2004, 316, 1065–1072. [Google Scholar] [CrossRef]
  158. Lee, I.O.; Lee, K.H.; Pyo, J.H.; Kim, J.H.; Choi, Y.J.; Lee, Y.C. Anti-Inflammatory Effect of Capsaicin in Helicobacter pylori Infected Gastric Epithelial Cells. Helicobacter 2007, 12, 510–517. [Google Scholar] [CrossRef]
  159. Shih, Y.-T.; Wu, D.-C.; Liu, C.-M.; Yang, Y.-C.; Chen, I.-J.; Lo, Y.-C. San-Huang-Xie-Xin-Tang inhibits Helicobacter pylori induced inflammation in human gastric epithelial AGS cells. J. Ethnopharmacol. 2007, 112, 537–544. [Google Scholar] [CrossRef]
  160. Miguel, G.; Faleiro, L.; Cavaleiro, C.; Salgueiro, L.; Casanova, J. Susceptibility of Helicobacter pylori to essential oil of Dittrichia viscosa subsp. revoluta. Phytother. Res. 2008, 22, 259–263. [Google Scholar] [CrossRef] [PubMed]
  161. Yang, J.-C.; Shun, C.-T.; Chien, C.-T.; Wang, T.-H. Effective prevention and treatment of Helicobacter pylori infection using a combination of catechins and sialic acid in AGS Cells and BALB/c mice. J. Nutr. 2008, 138, 2084–2090. [Google Scholar] [CrossRef] [PubMed]
  162. Gaus, K.; Huang, Y.; Israel, D.A.; Pendland, S.L.; Adeniyi, B.A.; Mahady, G.B. Standardized ginger (Zingiber officinale) extract reduces bacterial load and suppresses acute and chronic inflammation in Mongolian gerbils infected with CagA+ Helicobacter pylori. Pharm. Biol. 2009, 47, 92–98. [Google Scholar] [CrossRef] [PubMed]
  163. Pastene, E.; Speisky, H.; García, A.; Moreno, J.; Troncoso, M.; Figueroa, G. In vitro and in vivo effects of apple peel polyphenols against Helicobacter pylori. J. Agric. Food Chem. 2010, 58, 7172–7179. [Google Scholar] [CrossRef]
  164. Huang, H.-L.; Ko, C.-H.; Yan, Y.-Y.; Wang, C.-K. Antiadhesion and Anti-Inflammation Effects of Noni (Morinda citrifolia) fruit extracts on AGS cells during Helicobacter pylori infection. J. Agric. Food Chem. 2014, 62, 2374–2383. [Google Scholar] [CrossRef]
  165. Pastene, E.; Parada, V.; Avello, M.; Ruiz, A.; García, A. Catechin-based procyanidins from Peumus boldus Mol. aqueous extract inhibit Helicobacter pylori urease and adherence to adenocarcinoma gastric cells. Phytother. Res. 2014, 28, 1637–1645. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, X.; Gu, H.; Li, X.; Xu, Z.; Chen, Y.-S.; Li, Y. Anti-Helicobacter pylori compounds from the ethanol extracts of Geranium wilfordii. J. Ethnopharmacol. 2013, 147, 204–207. [Google Scholar] [CrossRef] [PubMed]
  167. Yakoob, J.; Jafri, W.; Mehmood, M.H.; Abbas, Z.; Tariq, K. Immunomodulatory effects of Psyllium extract on Helicobacter pylori interaction with gastric epithelial cells. J. Evid.-Based Complement. Altern. Med. 2016, 21, NP18–NP24. [Google Scholar] [CrossRef]
  168. Kouitcheu Mabeku, L.B.; Eyoum Bille, B.; Tchouangueu, T.F.; Nguepi, E.; Leundji, H. Treatment of Helicobacter pylori infected mice with Bryophyllum pinnatum, a medicinal plant with antioxidant and antimicrobial properties, reduces bacterial load. Pharm. Biol. 2016, 55, 603–610. [Google Scholar] [CrossRef]
  169. Zhang, Q.; Yue, L. Inhibitory Activity of Mangiferin on Helicobacter pylori-induced inflammation in human gastric carcinoma ags cells. Afr. J. Tradit. Complement. Altern. Med. 2016, 14, 263–271. [Google Scholar] [CrossRef]
  170. Li, C.; Huang, P.; Wong, K.; Xu, Y.; Tan, L.; Chen, H.; Lu, Q.; Luo, C.; Tam, C.; Zhu, L.; et al. Coptisine-induced inhibition of Helicobacter pylori: Elucidation of specific mechanisms by probing urease active site and its maturation process. J. Enzyme Inhib. Med. Chem. 2018, 33, 1362–1375. [Google Scholar] [CrossRef] [PubMed]
  171. Yen, C.-H.; Chiu, H.-F.; Huang, S.-Y.; Lu, Y.-Y.; Han, Y.-C.; Shen, Y.-C.; Venkatakrishnan, K.; Wang, C.-K. Beneficial effect of burdock complex on asymptomatic Helicobacter pylori-infected subjects: A randomized, double-blind placebo-controlled clinical trial. Helicobacter 2018, 23, e12469. [Google Scholar] [CrossRef] [PubMed]
  172. Kim, S.H.; Lim, J.W.; Kim, H. Astaxanthin prevents decreases in superoxide dismutase 2 level and superoxide dismutase ac-tivity in Helicobacter pylori-infected gastric epithelial cells. J. Cancer Prev. 2019, 24, 54–58. [Google Scholar] [CrossRef]
  173. Tian, J.; Si, X.; Wang, Y.; Gong, E.; Xie, X.; Zhang, Y.; Shu, C.; Li, B. Cyanidin-3-O-glucoside protects human gastric epithelial cells against Helicobacter pylori lipopolysaccharide-induced disorders by modulating TLR-mediated NF-κB pathway. J. Funct. Foods 2020, 68, 103899. [Google Scholar] [CrossRef]
  174. Goodman, C.; Lyon, K.N.; Scotto, A.; Smith, C.; Sebrell, T.A.; Gentry, A.B.; Bala, G.; Stoner, G.D.; Bimczok, D. A high-throughput metabolic microarray assay reveals antibacterial effects of black and red raspberries and blackberries against Helicobacter pylori infection. Antibiotics 2021, 10, 845. [Google Scholar] [CrossRef] [PubMed]
  175. Zhu, Y.; Liu, L.; Hu, L.; Dong, W.; Zhang, M.; Liu, Y.; Li, P. Effect of Celastrus orbiculatus in inhibiting Helicobacter pylori induced inflammatory response by regulating epithelial mesenchymal transition and targeting mir-21/pdcd4 signaling pathway in gastric epithelial cells. BMC Complement. Altern. Med. 2019, 19, 91. [Google Scholar] [CrossRef] [PubMed]
  176. Youssef, F.S.; Eid, S.Y.; Alshammari, E.; Ashour, M.L.; Wink, M.; El-Readi, M.Z. Chrysanthemum indicum and Chrysanthemum morifolium: Chemical composition of their essential oils and their potential use as natural preservatives with antimicrobial and antioxidant activities. Foods 2020, 9, 1460. [Google Scholar] [CrossRef]
  177. Ayoub, I.M.; Abdel-Aziz, M.M.; Elhady, S.S.; Bagalagel, A.A.; Malatani, R.T.; Elkady, W.M. Valorization of Pimenta racemosa essential oils and extracts: GC-MS and LC-MS phytochemical profiling and evaluation of Helicobacter pylori inhibitory activity. Molecules 2022, 27, 7965. [Google Scholar] [CrossRef]
  178. Kim, D.S.H.L.; Park, S.-Y.; Kim, J.-Y. Curcuminoids from Curcuma longa L. (Zingiberaceae) that protect pc12 rat pheochro-mocytoma and normal human umbilical vein endothelial cells from βa(1–42) insult. Neurosci. Lett. 2001, 303, 57–61. [Google Scholar] [CrossRef]
  179. Wu, X.; Li, X.; Dang, Z.; Jia, Y. Berberine demonstrates anti-inflammatory properties in Helicobacter pylori-infected mice with chronic gastritis by attenuating the Th17 response triggered by the b cell-activating factor. J. Cell. Biochem. 2018, 119, 5373–5381. [Google Scholar] [CrossRef] [PubMed]
  180. Wu, H.; Sun, Q.; Dong, H.; Qiao, J.; Lin, Y.; Yu, C.; Li, Y. Gastroprotective action of the extract of Corydalis yanhusuo in Helicobacter pylori infection and its bioactive component, dehydrocorydaline. J. Ethnopharmacol. 2023, 307, 116173. [Google Scholar] [CrossRef]
  181. Zhang, L.; Ma, J.; Pan, K.; Go, V.L.W.; Chen, J.; You, W. Efficacy of cranberry juice on Helicobacter pylori infection: A dou-ble-blind, randomized placebo-controlled trial. Helicobacter 2005, 10, 139–145. [Google Scholar] [CrossRef]
  182. Shmuely, H.; Yahav, J.; Samra, Z.; Chodick, G.; Koren, R.; Niv, Y.; Ofek, I. Effect of cranberry juice on eradication of Helicobacter pylori in patients treated with antibiotics and a proton pump inhibitor. Mol. Nutr. Food Res. 2007, 51, 746–751. [Google Scholar] [CrossRef] [PubMed]
  183. Chua, C.-S.; Yang, K.-C.; Chen, J.-H.; Liu, Y.-H.; Hsu, Y.-H.; Lee, H.-C.; Huang, S.-Y. The efficacy of blueberry and grape seed extract combination on triple therapy for Helicobacter pylori eradication: A randomised controlled trial. Int. J. Food Sci. Nutr. 2016, 67, 177–183. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, D.; Ke, L.; Ni, Z.; Chen, Y.; Zhang, L.-H.; Zhu, S.-H.; Li, C.-J.; Shang, L.; Liang, J.; Shi, Y.-Q. Berberine containing quadruple therapy for initial Helicobacter pylori eradication. Medicine 2017, 96, e7697. [Google Scholar] [CrossRef]
  185. Surh, Y.-J. Anti-tumor promoting potential of selected spice ingredients with antioxidative and anti-inflammatory activities: A short review. Food Chem. Toxicol. 2002, 40, 1091–1097. [Google Scholar] [CrossRef]
  186. Burger, O.; Weiss, E.; Sharon, N.; Tabak, M.; Neeman, I.; Ofek, I. Inhibition of Helicobacter pylori adhesion to human gastric mucus by a high-molecular-weight constituent of cranberry juice. Crit. Rev. Food Sci. Nutr. 2002, 42 (Suppl. S3), 279–284. [Google Scholar] [CrossRef] [PubMed]
  187. Gu, L.; Kelm, M.A.; Hammerstone, J.F.; Beecher, G.; Holden, J.; Haytowitz, D.; Gebhardt, S.; Prior, R.L. Concentrations of proanthocyanidins in common foods and estimations of normal consumption. J. Nutr. 2004, 134, 613–617. [Google Scholar] [CrossRef]
  188. Matsushima, M.; Suzuki, T.; Masui, A.; Kasai, K.; Kouchi, T.; Takagi, A.; Shirai, T.; Mine, T. Growth inhibitory action of cranberry on Helicobacter pylori. J. Gastroenterol. Hepatol. 2008, 23, S175–S180. [Google Scholar] [CrossRef]
  189. Malishev, R.; Shaham-Niv, S.; Nandi, S.; Kolusheva, S.; Gazit, E.; Jelinek, R. Bacoside-A, an Indian traditional-medicine sub-stance, inhibits β-amyloid cytotoxicity, fibrillation, and membrane interactions. ACS Chem. Neurosci. 2017, 8, 884–891. [Google Scholar] [CrossRef]
  190. Xu, T.-Z.; Shen, X.-Y.; Sun, L.-L.; Chen, Y.-L.; Zhang, B.-Q.; Huang, D.-K.; Li, W.-Z. Ginsenoside Rg1 protects against h2o2-induced neuronal damage due to inhibition of the NLRP1 inflammasome signalling pathway in hippocampal neurons in vitro. Int. J. Mol. Med. 2019, 43, 717–726. [Google Scholar] [CrossRef]
  191. Zolkiffly, S.Z.I.; Stanslas, J.; Abdul Hamid, H.; Mehat, M.Z. Ficus deltoidea: Potential inhibitor of pro-inflammatory mediators in lipopolysaccharide-induced activation of microglial cells. J. Ethnopharmacol. 2021, 279, 114309. [Google Scholar] [CrossRef]
  192. Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular mechanisms underlying protective effects of quercetin against mitochondrial dysfunction and progressive dopaminergic neurodegeneration in cell culture and mitopark transgenic mouse models of Parkinson’s disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef]
  193. Zhang, M.; Qian, C.; Zheng, Z.-G.; Qian, F.; Wang, Y.; Thu, P.M.; Zhang, X.; Zhou, Y.; Tu, L.; Liu, Q.; et al. Jujuboside A promotes Aβ clearance and ameliorates cognitive deficiency in Alzheimer’s disease through activating Axl/HSP90/PPARγ pathway. Theranostics 2018, 8, 4262–4278. [Google Scholar] [CrossRef]
  194. Xu, M.; Zhang, X.; Ren, F.; Yan, T.; Wu, B.; Bi, K.; Bi, W.; Jia, Y. Essential oil of Schisandra chinensis ameliorates cognitive decline in mice by alleviating inflammation. Food Funct. 2019, 10, 5827–5842. [Google Scholar] [CrossRef]
  195. Guan, L.; Mao, Z.; Yang, S.; Wu, G.; Chen, Y.; Yin, L.; Qi, Y.; Han, L.; Xu, L. Dioscin alleviates Alzheimer’s disease through regulating rage/nox4 mediated oxidative stress and inflammation. Biomed. Pharmacother. 2022, 152, 113248. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, Y.; Yang, X.; Wang, S.; Song, S. Ginsenoside Rg3 prevents cognitive impairment by improving mitochondrial dysfunction in the rat model of Alzheimer’s disease. J. Agric. Food. Chem. 2019, 67, 10048–10058. [Google Scholar] [CrossRef] [PubMed]
  197. Levites, Y.; Weinreb, O.; Maor, G.; Youdim, M.B.H.; Mandel, S. Green tea polyphenol (−)-epigallocatechin-3-gallate prevents n-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 2001, 78, 1073–1082. [Google Scholar] [CrossRef] [PubMed]
  198. Datla, K.P.; Christidou, M.; Widmer, W.W.; Rooprai, H.K.; Dexter, D.T. Tissue distribution and neuroprotective effects of citrus flavonoid tangeretin in a rat model of Parkinson’s disease. Neuroreport 2001, 12, 3871–3875. [Google Scholar] [CrossRef]
  199. Koh, S.-H.; Lee, S.M.; Kim, H.Y.; Lee, K.-Y.; Lee, Y.J.; Kim, H.-T.; Kim, J.; Kim, M.-H.; Hwang, M.S.; Song, C.; et al. The effect of epigallocatechin gallate on suppressing disease progression of ALS model mice. Neurosci. Lett. 2006, 395, 103–107. [Google Scholar] [CrossRef]
  200. Baluchnejadmojarad, T.; Roghani, M.; Nadoushan, M.R.J.; Bagheri, M. Neuroprotective effect of genistein in 6-hydroxydopamine hemi-parkinsonian rat model. Phytother. Res. 2009, 23, 132–135. [Google Scholar] [CrossRef]
  201. Kumar, P.; Kumar, A. Protective effect of hesperidin and naringin against 3-nitropropionic acid induced Huntington’s like symptoms in rats: Possible role of nitric oxide. Behav. Brain Res. 2010, 206, 38–46. [Google Scholar] [CrossRef]
  202. Pannangrong, W.; Wattanathorn, J.; Muchimapura, S.; Tiamkao, S.; Tong-un, T. Purple rice berry is neuroprotective and en-hances cognition in a rat model of Alzheimer’s disease. J. Med. Food 2011, 14, 688–694. [Google Scholar] [CrossRef]
  203. Oboh, G.; Ademiluyi, A.O.; Akinyemi, A.J. Inhibition of acetylcholinesterase activities and some pro-oxidant induced lipid peroxidation in rat brain by two varieties of ginger (Zingiber officinale). Exp. Toxicol. Pathol. 2012, 64, 315–319. [Google Scholar] [CrossRef]
  204. Gopinath, K.; Sudhandiran, G. Naringin modulates oxidative stress and inflammation in 3-nitropropionic acid-induced neu-rodegeneration through the activation of nuclear factor-erythroid 2-related factor-2 signalling pathway. Neuroscience 2012, 227, 134–143. [Google Scholar] [CrossRef] [PubMed]
  205. Sandhir, R.; Mehrotra, A. Quercetin supplementation is effective in improving mitochondrial dysfunctions induced by 3-nitropropionic acid: Implications in Huntington’s disease. Biochim. Biophys. Acta 2013, 1832, 421–430. [Google Scholar] [CrossRef] [PubMed]
  206. Arbabi, E.; Hamidi, G.; Talaei, S.A.; Salami, M. Estrogen agonist genistein differentially influences the cognitive and motor disorders in an ovariectomized animal model of parkinsonism. Iran. J. Basic Med. Sci. 2016, 19, 1285–1290. [Google Scholar] [CrossRef]
  207. El-Horany, H.E.; El-latif, R.N.A.; ElBatsh, M.M.; Emam, M.N. Ameliorative effect of quercetin on neurochemical and behav-ioral deficits in rotenone rat model of Parkinson’s disease: Modulating autophagy (quercetin on experimental Parkinson’s disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef] [PubMed]
  208. Huang, H.-J.; Chen, S.-L.; Chang, Y.-T.; Chyuan, J.-H.; Hsieh-Li, H.M. Administration of Momordica charantia enhances the neuroprotection and reduces the side effects of LiCl in the treatment of Alzheimer’s disease. Nutrients 2018, 10, 1888. [Google Scholar] [CrossRef]
  209. Singh, S.S.; Rai, S.N.; Birla, H.; Zahra, W.; Kumar, G.; Gedda, M.R.; Tiwari, N.; Patnaik, R.; Singh, R.K.; Singh, S.P. Effect of chlorogenic acid supplementation in MPTP-intoxicated mouse. Front. Pharmacol. 2018, 9, 757. [Google Scholar] [CrossRef]
  210. Zhou, T.; Zhu, M.; Liang, Z. (−)-Epigallocatechin-3-gallate modulates peripheral immunity in the MPTP-induced mouse model of Parkinson’s disease. Mol. Med. Rep. 2018, 17, 4883–4888. [Google Scholar] [CrossRef]
  211. Xian, Y.-F.; Mao, Q.-Q.; Wu, J.C.; Su, Z.-R.; Chen, J.-N.; Lai, X.-P.; Ip, S.-P.; Lin, Z.-X. Isorhynchophylline treatment improves the amyloid-β-induced cognitive impairment in rats via inhibition of neuronal apoptosis and tau protein hyperphosphorylation. J. Alzheimer’s Dis. 2014, 39, 331–346. [Google Scholar] [CrossRef] [PubMed]
  212. Ikram, M.; Jo, M.H.; Choe, K.; Khan, A.; Ahmad, S.; Saeed, K.; Kim, M.W.; Kim, M.O. Cycloastragenol, a triterpenoid saponin, regulates oxidative stress, neurotrophic dysfunctions, neuroinflammation and apoptotic cell death in neurodegenerative conditions. Cells 2021, 10, 2719. [Google Scholar] [CrossRef]
  213. Zhang, S.; Tomata, Y.; Sugiyama, K.; Sugawara, Y.; Tsuji, I. Citrus consumption and incident dementia in elderly Japanese: The Ohsaki cohort 2006 study. Br. J. Nutr. 2017, 117, 1174–1180. [Google Scholar] [CrossRef]
  214. Heo, J.-H.; Lee, S.-T.; Chu, K.; Oh, M.J.; Park, H.-J.; Shim, J.-Y.; Kim, M. An open-label trial of Korean red ginseng as an ad-juvant treatment for cognitive impairment in patients with Alzheimer’s disease. Eur. J. Neurol. 2008, 15, 865–868. [Google Scholar] [CrossRef]
  215. Lee, S.-T.; Chu, K.; Sim, J.-Y.; Heo, J.-H.; Kim, M. Panax ginseng enhances cognitive performance in Alzheimer disease. Alz-heimer Dis. Assoc. Disord. 2008, 22, 222–226. [Google Scholar] [CrossRef] [PubMed]
  216. Cox, K.H.; Pipingas, A.; Scholey, A.B. Investigation of the effects of solid lipid curcumin on cognition and mood in a healthy older population. J. Psychopharmacol. 2014, 29, 642–651. [Google Scholar] [CrossRef] [PubMed]
  217. Kean, R.J.; Lamport, D.J.; Dodd, G.F.; Freeman, J.E.; Williams, C.M.; Ellis, J.A.; Butler, L.T.; Spencer, J.P. Chronic consumption of flavanone-rich orange juice is associated with cognitive benefits: An 8-wk, randomized, double-blind, placebo-controlled trial in healthy older adults. Am. J. Clin. Nutr. 2015, 101, 506–514. [Google Scholar] [CrossRef]
  218. Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef]
  219. Alharbi, M.H.; Lamport, D.J.; Dodd, G.F.; Saunders, C.; Harkness, L.; Butler, L.T.; Spencer, J.P.E. Flavonoid-rich orange juice is associated with acute improvements in cognitive function in healthy middle-aged males. Eur. J. Nutr. 2015, 55, 2021–2029. [Google Scholar] [CrossRef] [PubMed]
  220. Park, S.-Y.; Kim, D.S.H.L. Discovery of natural products from Curcuma longa that protect cells from beta-amyloid insult:  a drug discovery effort against Alzheimer’s disease. J. Nat. Prod. 2002, 65, 1227–1231. [Google Scholar] [CrossRef] [PubMed]
  221. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin inhibits formation of amyloid β oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2004, 280, 5892–5901. [Google Scholar] [CrossRef]
  222. Ogunruku, O.O.; Oboh, G.; Passamonti, S.; Tramer, F.; Boligon, A.A. Capsicum annuum var. grossum (Bell Pepper) inhibits β-secretase activity and β-amyloid1–40 aggregation. J. Med. Food 2017, 20, 124–130. [Google Scholar] [CrossRef]
  223. Sengupta, U.; Nilson, A.N.; Kayed, R. The role of amyloid-β oligomers in toxicity, propagation, and immunotherapy. EBio-Medicine 2016, 6, 42–49. [Google Scholar] [CrossRef]
  224. Daubner, S.C.; Le, T.; Wang, S. Tyrosine hydroxylase and regulation of dopamine synthesis. Arch. Biochem. Biophys. 2011, 508, 1–12. [Google Scholar] [CrossRef] [PubMed]
  225. Zhu, Y.; Zhang, J.; Zeng, Y. Overview of tyrosine hydroxylase in Parkinson’s disease. CNS Neurol. Disord. Drug Targets 2012, 11, 350–358. [Google Scholar] [CrossRef]
  226. Compagnoni, G.M.; Di Fonzo, A.; Corti, S.; Comi, G.P.; Bresolin, N.; Masliah, E. The role of mitochondria in neurodegenerative diseases: The lesson from Alzheimer’s disease and Parkinson’s disease. Mol. Neurobiol. 2020, 57, 2959–2980. [Google Scholar] [CrossRef]
  227. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef]
  228. Sas, K.; Robotka, H.; Toldi, J.; Vécsei, L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J. Neurol. Sci. 2007, 257, 221–239. [Google Scholar] [CrossRef] [PubMed]
  229. Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Grigoriadis, N.; Deretzi, G.; Tzilves, D.; Katsinelos, P.; Tsolaki, M.; Chatzopoulos, D.; et al. Eradication of Helicobacter pylori may be beneficial in the management of Alzheimer’s disease. J. Neurol. 2009, 256, 758–767. [Google Scholar] [CrossRef] [PubMed]
  230. Chang, Y.-P.; Chiu, G.-F.; Kuo, F.-C.; Lai, C.-L.; Yang, Y.-H.; Hu, H.-M.; Chang, P.-Y.; Chen, C.-Y.; Wu, D.-C.; Yu, F.-J. Eradication of Helicobacter pylori is associated with the progression of dementia: A population-based study. Gastroenterol. Res. Pract. 2013, 2013, e175729. [Google Scholar] [CrossRef] [PubMed]
  231. Fasano, A.; Visanji, N.P.; Liu, L.W.C.; Lang, A.E.; Pfeiffer, R.F. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2015, 14, 625–639. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic diagram of H. pylori virulence factor.
Figure 1. Schematic diagram of H. pylori virulence factor.
Molecules 28 07150 g001
Figure 2. APP proteolytic amyloidogenic and non-amyloidogenic pathway.
Figure 2. APP proteolytic amyloidogenic and non-amyloidogenic pathway.
Molecules 28 07150 g002
Figure 3. Possible relationship between H. pylori infection and neurodegenerative disease.
Figure 3. Possible relationship between H. pylori infection and neurodegenerative disease.
Molecules 28 07150 g003
Figure 4. Anti-H. pylori activity of phytochemicals from natural sources.
Figure 4. Anti-H. pylori activity of phytochemicals from natural sources.
Molecules 28 07150 g004
Figure 5. Neuroprotection activity of different phytochemical from natural sources.
Figure 5. Neuroprotection activity of different phytochemical from natural sources.
Molecules 28 07150 g005
Table 1. Assessment of anti-H. pylori activity from natural sources in in vitro studies.
Table 1. Assessment of anti-H. pylori activity from natural sources in in vitro studies.
Test MaterialActivityFindingsSource
Ginger (Gingerol)Inhibit H. pylori growthInhibit growth of CagA+ H. pylori strains (MIC: 6.25–50 µg/mL)[156]
Curcuma longa L. (Curcumin)Anti-inflammatory propertiesMolecules 28 07150 i001 IκBα degradation (up to 80 µM)
Molecules 28 07150 i002 IKKα and β activity (up to 80 µM)
Molecules 28 07150 i003 NF-κB DNA-binding (up to 80 µM)
[157]
Chilli pepper (Capsaicin)Anti-inflammatory propertiesMolecules 28 07150 i004 H. pylori-induced IL-8 production in MKN45 and AGS cell (100 µM capsaicin, 43.2% and 70%, respectively, compared to control)
Molecules 28 07150 i005 IL-8 mRNA expression (100 µM capsaicin)
Molecules 28 07150 i006 Reduce H. pylori NF-κB activation (100 µM capsaicin)
[158]
San-Huang-Xie-Xin-Tang (Coptis chinesis Franch, Scutellaria baicalensis Georgi, and Rheum officinale Baill) (Baicalin)Anti-inflammatory propertiesMolecules 28 07150 i007 H. pylori induced COX-2 enhancement (treatment vs. control group, p < 0.05)
Molecules 28 07150 i008 IκBα degradation and nuclear translocation of NF-κB p50 subunit (treatment vs. control group, p < 0.05)
Molecules 28 07150 i009 iNOS and IL-8 mRNA expression (treatment vs. control group, p < 0.05)
Molecules 28 07150 i010 decreased NO and IL-8 production (treatment vs. control group, p < 0.05)
[159]
Dittrichia viscosa subsp. Revoluta
(Essential oil (3-methoxy cuminyl isobutyrate, α-cadinol and α-eudesmol)
Inhibit H. pylori growthEssential oil derived from Dittrichia viscosa especially fraction 5 and 7 show highest anti-H. pylori activity [160]
Green tea (Catechin and pure sialic acid)Antioxidant propertiesMolecules 28 07150 i011 Reduce O2, H2O2 count, NO production (treatment vs control group, p < 0.05)[161]
Anti-inflammatory propertiesMolecules 28 07150 i012 iNOS expression
Anti-apoptosisMolecules 28 07150 i013 Inhibited apoptosis and reduced apoptosis related protein expression (treatment vs. control group, p < 0.05)
Ginger (Gingerol)Anti-inflammatory propertiesMolecules 28 07150 i014 COX-2 (IC50: 8.5 µg/mL)
Molecules 28 07150 i015 NF-κB transcription (IC50: 24.6 µg/mL)
Molecules 28 07150 i016 Inflammatory cytokine production (IL-1β, IL-6, IL-8, TNF-α (IC50: 3.89, 7.7, 8.5, and 8.37 µg/mL respectively))
[162]
Apple peel polyphenolAnti-apoptosisMolecules 28 07150 i017 H. pylori stimulated vacuolation in HeLa cell (IC50: 390 µg GAE/mL)[163]
Anti-adhesion propertiesMolecules 28 07150 i018 60% adhesion at concentration 5 mg GAE/mL
Noni fruitAnti-adhesion propertiesMolecules 28 07150 i019 Adhesion of H. pylori to AGS cell (treatment vs infected group, p < 0.05)
Molecules 28 07150 i020 Intracellular CagA level (treatment vs infected group, p < 0.05)
[164]
Anti-inflammatory propertiesMolecules 28 07150 i021 Inflammatory markers (IL-8, iNOS, COX-2) and neutrophil chemotaxis (treatment vs. infected group, p < 0.05)
Peumus boldus Mol. (Catechin)Inhibit urease activityMolecules 28 07150 i022 Urease activity from H. pylori [165]
Anti-adhesion propertiesMolecules 28 07150 i023 Adhesion ratio of H. pylori to AGS cell (treatment vs. infected group, p < 0.05)
Geranium wilfordii (Corilagin and 1,2,3,6-tetra-O-galloyl-b-D-glucose)Inhibit H. pylori growthEthanol and ethyl acetate extract inhibited H. pylori growth (MIC: 40 and 30 μg/mL, respectively) [166]
Plantago ovataAnti-inflammatory propertiesMolecules 28 07150 i024 Basal and H. pylori-stimulated IL-8 secretion up to 74.51% and 66.67%, respectively (p < 0.001)
Molecules 28 07150 i025 CagA-positive H pylori–induced IL-8 mRNA expression up to 67.6% (p < 0.0001)
Molecules 28 07150 i026 Nf-κB activation (p = 0.0001)
[167]
Bryophyllum pinnatumInhibit H. pylori growthBryophyllum pinnatum methanol extract showed anti-H. pylori activity (MIC: 32 μg/mL and MBC: 256 μg/mL)[168]
Mangiferin indica (Mangiferin)Inhibit H. pylori growthMolecules 28 07150 i027 Growth of H. pylori (dose dependent, up to 100 μg/mL, p < 0.05)[169]
Anti-adhesion propertiesMolecules 28 07150 i028 H. pylori adhesion to AGS cell (p < 0.05, treatment vs. control group)
Anti-inflammatory propertiesMolecules 28 07150 i029 Inflammatory cytokines (NF-κB p65 sub-unit, TNF-α, IL-1β, and IL-8) (p < 0.01) and enzyme expression (COX-2, iNOS) (p < 0.05)
Coptis chinensis Franch (Berberine, palmatine, coptisine, jatrorrhizine, and epiberberine)Inhibit H. pylori growthCoptisine showed the highest anti-H. pylori activity with MIC and MBC 25 to 50 μg/mL and 37.5 to 125 μg/mL, respectively[170]
Inhibit urease activityInhibit urease activity and maturation
Burdock complex
(Arctium lappa, Angelica sinensis, Lithospermum erythrorhizon, and Sesamum indicum oil)
Anti-adhesion propertiesMolecules 28 07150 i030 Adhesion of H. pylori to AGS cell (p < 0.05, compared with H. pylori-infected group)[171]
Anti-inflammatory propertiesMolecules 28 07150 i031 Inflammatory marker (IL-8, TNF-α) (p < 0.05, compared with H. pylori-infected group)
AstaxanthinAntioxidant propertiesPrevent the SOD2 level decrease and increase SOD activity, and mitochondrial ROS production in AGS cell [172]
Blueberry (Cyanindin-3-O-Glucoside)Anti-inflammatory propertiesC3G from blueberry suppressed abnormal DNA synthesis, inflammation, and TLR2 and TLR4 expression; induced apoptosis; and deactivated TLR-mediated NF-κB signaling in LPS-treated cell[173]
Black raspberry (Anthocyanin)Inhibit H. pylori growthInhibited growth of H. pylori without having side effects on AGS cell (MIC: 5 µg/mL)[174]
Celastrus orbiculatusAnti-inflammatory propertiesReduces inflammatory response by regulating epithelial–mesenchymal transition; suppressed methylation of PDCD4 promoter and inhibited microRNA-21, thus enhancing the PDCD4 expression [175]
Chrysanthemum indicum and Chrysanthemum morifolium (Essential oil (major constituent camphor))Inhibit H. pylori growthBoth essential oil of C. indicum and C. morifolium showed potent anti-H. pylori activity with IC50 3.63 and 3.78 µg/mL, respectively[176]
Pimenta racemosa (leaves and stem essential oil (eugenol) and methanolic extract)Inhibit H. pylori growthPimenta racemosa stem essential oil showed the highest anti-H. pylori activity compared to others with MIC: 3.9 μg/mL and it inhibited H. pylori urease activity simulated with in silico molecular modelling [177]
Molecules 28 07150 i032 Indicating decrease in the issue.
Table 2. Assessment of anti-H. pylori activity from natural sources in in vivo studies.
Table 2. Assessment of anti-H. pylori activity from natural sources in in vivo studies.
Test MaterialSubjectActivityFindingsSource
Green tea (Catechin and sialic acid)Male BALB/c miceAnti-inflammatory propertiesPre-treatment and post-treatment with catechin and/or sialic acid significantly reduced H. pylori infection, mucosal damage, and gastritits score (treatment vs. control group, p < 0.05)[161]
Ginger (Gingerol)Mongolian gerbilsAnti-inflammatory propertiesSignificantly reduces mucosal and submucosal inflammation, cryptitis, epithelial degeneration, and erosion due to H. pylori infection compared to control[178]
Polyphenol rich apple peel extractC57BL6/J miceAnti-adhesion propertiesAdministration of apple peel polyphenol could reduce adhesion of H. pylori;
reduced inflammation, lowering malonaldehyde levels and gastritis score in mice
[163]
Anti-inflammatory properties
Bryophyllum pinnatumSwiss miceInhibit H. pylori growthBryophyllum pinnatum significantly reduced bacterial colonization in gastric tissue and bacterial load in Swiss mice[168]
BerberineMale C57Bl/6 miceAnti-inflammatory propertiesBerberine treatment suppressed pro inflammatory cytokines and upregulated anti-inflammatory cytokines expression [179]
Corydalis yanhusuo (Benzylisoquinoline alkaloids)Male miceInhibit H. pylori growthTwo different extracts of Corydalis yanhusuo (ethanol and chloroform) inhibited the growth of H. pylori, with MIC ranging from 50 to 100 μg/mL and MBC ranging from 100 to 200 μg/mL; chloroform extract of Corydalis yanhusuo reduces survival ability of H. pylori in gastric mucosa and repairs gastric damage together with reduction of H. pylori IgG in infected mice[180]
Cranberry (A-type proanthocyanidin)H. pylori-positive adultsAnti-adhesion propertiesConsumption of cranberry juice could significantly reduce H. pylori infection compared to placebo group[181]
Cranberry (A-type proanthocyanidin)H. pylori-positive adultsAnti-adhesion propertiesCranberry juice addition to standard triple therapy (Omeprazole, Amoxicillin, and Clarithromycin) could significantly improve H. pylori eradication rates in female subjects [182]
Cranberry (A-type proanthocyanidin) and Lactobacillus johnsonii La1Asymptomatic H. pylori-positive childrenAnti-adhesion propertiesCombination of cranberry juice and L. johnsonii La1 reduced H. pylori infection compared to each test material alone and control group, but no synergistic inhibitory effect observed[24]
Blueberry and grape seed extract (Proanthocyanidin)H. pylori-positive patientAntioxidant propertiesCombination of blueberry and grape seed extract did not produce a significant change in eradication rate of H. pylori compared to placebo group[183]
BerberineH. pylori-positive patientAntioxidant propertiesNo significant difference between berberine containing quadruple therapy eradication rate and adverse effect compared to bismuth containing quadruple therapy[184]
Burdock complex
(Arctium lappa, Angelica sinensis, Lithospermum erythrorhizon, and Sesamum indicum oil)
Asymptomatic H. pylori-positive subjectAnti-adhesion propertiesSignificantly reduced UBT value (compared to placebo, p < 0.05)[171]
Anti-inflammatory propertiesSignificantly reduced inflammatory marker and (compared to placebo, p < 0.05)
Antioxidant propertiesImproved antioxidant status and plasma phenolic level (compared to placebo, p < 0.05) and heal the ulcer in the stomach
Cranberry (A-type proanthocyanidin)H. pylori positive adultsAnti-adhesion propertiesConsumption of high-proanthocyanidin cranberry juice twice a day (44 mg/serving) for 8 weeks could significantly decrease H. pylori infection compared to placebo; consumption of encapsulated cranberry powder not significantly effective to reduce H. pylori infection[20]
Table 3. Assessment of neuroprotective activity of phytochemical from natural sources in in vitro studies.
Table 3. Assessment of neuroprotective activity of phytochemical from natural sources in in vitro studies.
Test MaterialCell lineActivityFindingsSource
Curcuma longa L. (Curcumin, demethoxycurcumin, and bisdemethoxycurcumin)PC12 cells and human umbilical vein endothelial cells (HUVEC)Anti-apoptosis activityThree curcuminoids from Curcuma longa L. found to protect PC12 cells and HUVEC from Aβ insult [162]
Curcuma longa L. (9 different isolated compounds)PC12 cellsAnti-apoptosis activityFive isolated compounds from Curcuma longa L. effectively protected PC12 cells from Aβ cytotoxicity [163]
Capsicum annuum var. grossum
(Polyphenol rich extract)
In vitro studyReduce Aβ aggregationPhenolic extract from bell pepper could counteract initial aggregation of Aβ and prevent further aggregation (fibril formation)[164]
Bacopa monnieri (Bacoside-A)SH-SY5Y cellsAnti-apoptosis activityReduced cell cytotoxicity and inhibited fibril formation both in buffer solution only and in the presence of membrane vesicles[189]
Ginseng (Ginsenoside Rg1)Primary hippocampal neuronsAnti-inflammatory propertiesGinsenoside Rg1 reduced ROS production, NOX2, and NLRP1 inflammasome due to H2O2 treatment. [190]
Anti-apoptosis activityGinsenoside Rg1 also reduced apoptosis, activation of β-galactosidase, and neuronal damage after H2O2 treatment.
Ficus deltoidea Jack (Vitexin and isovitexin)Mouse microglial (BV-2) cells Anti-inflammatory propertiesTreatment with Ficus deltoidea Jack extract significantly reduced ROS, NO, TNF-α, IL-1β, and IL-6 production from microglial cell after treatment with LPS[191]
QuercetinMN9D dopaminergic neuronal cells Improve mitochondria functionIncreased mitochondrial biogenesis and bioenergetics capacity of MN9D cell and reduced 6-hydroxydopmaine induced toxicity[192]
Semen ziziphi spinosae (Jujuboside A)BV-2 cellsAnti-apoptosis activityJuA treatment upregulated expression of HSP90β, preserved PPARγ levels, promoted interaction between HSP90β and PPARγ, and promoted the clearance of Aβ42[193]
Schisandra chinensis (Essential oil)BV-2 cells Anti-inflammatory propertiesSchisandra chinensis essential oil treatment decreased NO production and blocked MAPK activation in LPS-stimulated BV-2 microglial cell[194]
Dioscoreae nipponicae
(Dioscin)
SH-SY5Y cell Anti-apoptosis activityDioscin improved cell viability [195]
Antioxidant activityReduce ROS production due to H2O2 injury in SH-SY5Y cell line
Table 4. Assessment of neuroprotective activity of phytochemical from natural sources in in vivo studies.
Table 4. Assessment of neuroprotective activity of phytochemical from natural sources in in vivo studies.
Test MaterialCompound SubjectFindingsSource
GinsengGinsenoside Rg3Male Wistar ratsGinsenoside Rg3 significantly reduced neuronal apoptosis and apoptosis related protein after treatment of D-galactose; ginsenoside Rg3 also improved antioxidant status and mitochondrial function in D-galactose-induced AD rats [196]
Green tea extract(−) Epigallocatechin-3-gallate Male C57/BL miceGreen tea extract treatment reduced N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity and prevented dopaminergic neuronal loss[197]
CitrusTangeretinMale Sprague-Dawley ratsTangeretin can cross the blood–brain barrier and protect neuronal cells against 6-OHDA toxicity [198]
Epigallocatechin gallateTransgenic mice carrying human G93A mutated SOD1 geneEGCG treatment prolonged lifespan and the symptoms onset and increased the survival rate of experimental mice[199]
GenisteinMale Sprague-Dawley ratsHigh dose genistein treatment showed neuroprotective effect against 6-OHDA toxicity[200]
Hesperidin and naringinWistar ratsPre-treatment with hesperidin and naringin reduced behavioral alteration, oxidative stress, and mitochondrial enzyme dysfunction; this effect was further enhanced when combined with NOS inhibitor (L-NAME) [201]
Oryza sativa (Rice berry, purple)AnthocyaninWistar ratsPrevented memory impairment and hippocampal neurodegeneration; decreased AChE activity and lipid peroxidation [202]
Zingiber officinale (Red and White Ginger) Wistar strain albino ratsBoth extracts inhibited AChE individually and combined together, and both extracts significantly decreased the SNP and QA elevated brain MDA contents[203]
NaringinMale Wistar ratsImprovement of glutathione/oxidized glutathione ratio and reduced free radical level due to 3-nitropropionic acid treatment through Nrf2 activation [204]
QuercetinFemale Wistar ratsQuercetin treatment improved mitochondrial function and antioxidant enzymes, as well as reducing astrogliosis and neurobehavioral deficits in experimental rats[205]
GenisteinFemale Wistar ratsImprovement in Morris water maze result and neuroprotective effect on dopaminergic neuronal cells[206]
QuercetinAlbino ratsSignificant reduction of behavioral impairment due to rotenone; reduced endoplasmic reticulum stress-induced apoptosis and oxidative stress[207]
QuercetinMitoPark transgenic miceImproved behavioral change, and reduced dopamine depletion and neuronal loss in MitoPark transgenic mice[192]
Momordica charantia C57BL/6J and 3 × Tg-AD micePrevent memory deficits; reduced neuronal loss, gliosis, Aβ level, and tau hyperphosporylation; and increased synaptic-related protein and pS9-GSK3β expression[208]
Chlorogenic acidSwiss albino male miceChlorogenic acid significantly improve motor coordination and antioxidant status. Chlorogenic acid also reduce neuroinflammation and inhibit release of proinflammatory cytokines[209]
(−) Epigallocatechin-3-gallateC57BL/6J miceImprovement in movement behavior and protection of tyrosine hydroxylase (+) cells against MPTP toxicity, increased CD3+/CD4+ and CD3+/CD8+ T lymphocyte ratio, and reduced pro-inflammatory cytokine production[210]
Uncaria rhynchophyllaIsorhynchophylline (IRN)Male Sprague-Dawley ratsIRN treatment alleviated cognitive decline due to Aβ25-35, reduced neuronal apoptosis, and suppressed tau hyperphosphorylation; additionally, IRN also inhibited GSK-3β activity and activated PI3K phosphorylation, which play a role in neuroprotection[211]
Semen ziziphi spinosaeJujuboside AAPP/PS1 transgenic miceJuA significantly reduced cognitive deficiency in APP/PS1 transgenic mice, and significantly reduced soluble Aβ42 levels and plaque numbers in the brain[193]
Schisandra chinensisEssential oil Male KM miceSchisandra chinensis essential oil can improve cognitive decline in mice, suppressed pro-inflammatory cytokines, and inhibited p38 activation in the mice model[194]
Astragalus radixCycloastragenolC57BL/6N miceCycloastragenol upregulated the expression of Nrf2, HO-1, p-TrKB, BDNF, and NeuN and downregulated the expression of p-JNK, p-P-38, and p-Erk; cycloastragenol reduced the activated microglia, inflammatory cytokines, apoptosis, and memory dysfunction[212]
Dioscoreae nipponicae DioscinC57BL/6 miceResult from in vivo study showed dioscin improved spatial learning and memory; restored MDA, Aβ42, AChE, ACh, and SOD levels; and restored brain histopathological change; dioscin downregulated the expression of RAGE and NOX4 and upregulated Nrf2 and HO-1; dioscin also downregulated the levels of p-NF-κB(p-p65)/NF-κB(p65), AP-1, and inflammatory factors [195]
Citrus Men and women aged ≥65, living in Ohsaki City, JapanFrequent consumption of citrus associated with lower risk of getting dementia [213]
Korean Red Ginseng (KRG)High KRG dose (9 g/day), low KRG dose (4.5 g/day), control for 12 weeks intervention61 patients with ADHigh dose KRG significantly improved Alzheimer’s Diseases Assessment Scale (ADAS) and Clinical Dementia Rating (CDR) compared to control; KRG group showed improvement on Mini Mental Status Examination (MMSE) but no significant difference with the control group[214]
Panax GinsengPanax Ginseng powder (4.5 g/day) and control for 12 weeks97 patients with probable AD by NINDS-ADRDA criteria Baseline MMSE and ADAS showed no difference between 2 groups; after intervention for 12 weeks, the group treated with panax ginseng showed MMSE and ADAS score improvement and after discontinuation of panax ginseng, MMSE and ADAS score declined to the level of the control group [215]
Cherry juiceAnthocyanin (200 mL of cherry juice/day for 12 weeks)Elder adult (age 70+) with mild to moderate dementiaSignificantly improved verbal fluency, short-term and long-term memory, and reduction of systolic and diastolic blood pressure, but no alteration of inflammation markers[22]
Curcumin Healthy adults Curcumin administration significantly improved sustained attention and working memory tasks compared to placebo; working memory and mood were significantly better after chronic treatment compared to placebo; curcumin treatment also significantly reduced total and LDL cholesterol[216]
CocoaFlavonol (1 dose daily for 8 weeks)
  • ≈990 mg/day (high),
  • ≈520 mg/day (intermediate)
  • ≈45 mg/day (low)
Elder people with mild cognitive impairment Time required to complete cognitive and verbal tests was significantly lower in the high and intermediate flavonol groups, compared to the low flavonol group[21]
Orange Juice
  • High flavanone (305 mg) and low flavanone (37 mg) daily for 8 weeks
Healthy older adultsHigh flavanone orange juice gives better improvement on global cognition score compared to the low flavanone group; no significant effect observed of flavanone consumption on mood changes[217]
Resveratrol (500 mg/day of Resveratrol (with dose escalation by 500 mg increments every 13 weeks))People aged > 45 with:
  • Diagnosed with probable AD
  • Mini-Mental State Examination (MMSE) score 14–26
  • Modified Hachinski Score < 5
stable use of cholinesterase inhibitors or memantine
Resveratrol was safe and well tolerated and some alteration of AD biomarkers were observed but a further and bigger study is needed to find evidence[218]
Orange juice Flavonoid-rich orange juice (272 mg/240 mL) or calorie-matched placebo Males aged 30–65 years oldFlavonoid-rich orange juice improved cognitive function, psychomotor speed, and subjective alertness compared to placebo[219]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tandoro, Y.; Chen, B.-K.; Ali, A.; Wang, C.-K. Review of Phytochemical Potency as a Natural Anti-Helicobacter pylori and Neuroprotective Agent. Molecules 2023, 28, 7150. https://doi.org/10.3390/molecules28207150

AMA Style

Tandoro Y, Chen B-K, Ali A, Wang C-K. Review of Phytochemical Potency as a Natural Anti-Helicobacter pylori and Neuroprotective Agent. Molecules. 2023; 28(20):7150. https://doi.org/10.3390/molecules28207150

Chicago/Turabian Style

Tandoro, Yohanes, Bo-Kai Chen, Asif Ali, and Chin-Kun Wang. 2023. "Review of Phytochemical Potency as a Natural Anti-Helicobacter pylori and Neuroprotective Agent" Molecules 28, no. 20: 7150. https://doi.org/10.3390/molecules28207150

Article Metrics

Back to TopTop