Next Article in Journal
The Effect of SGLT2 Inhibitors on the Development of Contrast-Induced Nephropathy in Diabetic Patients with Non-ST Segment Elevation Myocardial Infarction
Previous Article in Journal
The Predictive Value of Monocyte/High-Density Lipoprotein Ratio (MHR) and Positive Symptom Scores for Aggression in Patients with Schizophrenia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Controlling the Impact of Helicobacter pylori-Related Hyperhomocysteinemia on Neurodegeneration

by
Jannis Kountouras
1,*,
Michael Doulberis
1,2,
Apostolis Papaefthymiou
1,3,4,
Stergios A. Polyzos
4,
Christos Zavos
1,
Evangelos Kazakos
1,5,
Stergios Arapoglou
1,6,
Foteini Kyrailidi
1,
Maria C. Mouratidou
1,
Marina Boziki
7 and
Elisabeth Vardaka
1,8
1
Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Macedonia, Greece
2
Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
3
Pancreaticobiliary Medicine Unit, University College London Hospitals (UCLH), London W1W 6DN, UK
4
First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
5
School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, 50100 Kozani, Macedonia, Greece
6
Fifth Surgical Department, Medical School, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Macedonia, Greece
7
2nd Neurology Department, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, 54124 Thessaloniki, Macedonia, Greece
8
Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, 57400 Thessaloniki, Macedonia, Greece
*
Author to whom correspondence should be addressed.
Medicina 2023, 59(3), 504; https://doi.org/10.3390/medicina59030504
Submission received: 24 December 2022 / Revised: 27 February 2023 / Accepted: 28 February 2023 / Published: 4 March 2023
(This article belongs to the Section Neurology)

Abstract

:
Helicobacter pylori infection consists a high global burden affecting more than 50% of the world’s population. It is implicated, beyond substantiated local gastric pathologies, i.e., peptic ulcers and gastric cancer, in the pathophysiology of several neurodegenerative disorders, mainly by inducing hyperhomocysteinemia-related brain cortical thinning (BCT). BCT has been advocated as a possible biomarker associated with neurodegenerative central nervous system disorders such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and/or glaucoma, termed as “ocular Alzheimer’s disease”. According to the infection hypothesis in relation to neurodegeneration, Helicobacter pylori as non-commensal gut microbiome has been advocated as trigger and/or mediator of neurodegenerative diseases, such as the development of Alzheimer’s disease. Among others, Helicobacter pylori-related inflammatory mediators, defensins, autophagy, vitamin D, dietary factors, role of probiotics, and some pathogenetic considerations including relevant involved genes are discussed within this opinion article. In conclusion, by controlling the impact of Helicobacter pylori-related hyperhomocysteinemia on neurodegenerative disorders might offer benefits, and additional research is warranted to clarify this crucial topic currently representing a major worldwide burden.

1. Introduction

Recent data indicate the implications of microorganisms in the pathophysiology of neurodegeneration [1,2]. For instance, according to the Alzheimer’s disease (AD) pathogen theory, microorganisms operate as triggers, in conjunction with genetic variables, in starting the accumulation and/or processing of amyloid-β (Aβ), hyperphosphorylated tau proteins, and inflammation in the brains of AD [3]. Furthermore, brain cortical thinning (BCT), a hallmark feature mainly of brain aging, is linked with cognitive decline [4] and memory function disturbances [5]. BCT has been identified in many neurodegenerative disorders including AD [6], Parkinson’s disease (PD) [7], multiple sclerosis [8], or schizophrenia [9]. It is used as a biomarker of cognitive performance [10] to recognize mild cognitive-impairment subtypes [11]; to follow the clinical progression of disorders such as PD [7] or the progression of mild cognitive impairment to AD [10]; and to evaluate the effectiveness of interventional and rehabilitation procedures targeting improvements in sensory, motor, and cognitive functions [11].
Specifically, BCT, among other radiological MRI measurements, has been advocated as a possible biomarker associated with neurodegeneration either for primary neurodegenerative central nervous system (CNS) disorders such as AD [12] and/or PD [13] or other CNS disorders with a neurodegenerative pathological component, such as multiple sclerosis [13] and/or cardio-cerebrovascular-related dementia [14,15]. There are currently no diagnostic criteria for BCT; rather, longitudinal studies in the field provide estimates of normative rates in CNS pathology compared to age-related cortical thinning [16,17]. T1-weighted three-dimensional structural MRI scans are frequently used for this purpose, although advanced neuroimaging methodologies are continuously evolving [16].
According to the infection hypothesis in relation to neurodegeneration, microorganisms have been advocated as triggers and/or mediators of neurodegenerative diseases such as the development of AD [18]. The infection hypothesis of AD proposes that chronic infection with viral, bacterial, and/or fungal pathogens might be a trigger for AD onset during aging—probably via inflammatory processes—the risk of which seems predominantly high in apolipoprotein E (APOE) ε44 allele carriers. More specifically, proposed pathogens include latent neurotropic viruses (Herpes simplex virus type-1 and type-2; Human herpes- virus 4, 5, 6, and 7), gastrointestinal bacteria (Helicobacter pylori), periodontal bacteria (e.g., Porphyromonas gingivalis), pulmonary bacteria (e.g., Chlamydophila pneumoniae), spirochetes (e.g., Borrelia burgdorferi), and other infectious pathogens [18,19,20,21]. Over the last years, many detailed reviews have summarized the evidence about herpes simplex virus type-1, Chlamydophila pneumoniae, Helicobacter pylori, Porphyromonas gingivalis, and other oral infectious agents with the pathophysiology of AD [19].
In this regard, Helicobacter pylori (H. pylori) infection consists one of the most common chronic infections since it affects more than 50% of the world population [22]. Emerging evidence supports its involvement in the pathophysiology of many neurodegenerative disorders [21,23,24,25,26,27,28]. Specifically, found in gastric mucosa of ≥50% of humans worldwide, H. pylori can infect children, becoming chronic during adulthood if untreated; in most patients, H. pylori infections are acquired in childhood and, if untreated, persist throughout life [29], causing the chronic inflammatory process of the stomach lining and leading to many gastric as well as other upper and lower gastrointestinal and extra-gastrointestinal pathologies [30,31,32,33]. Therefore, epidemiological estimates of H. pylori prevalence refer to the untreated chronic H. pylori infection in adulthood. In this regard, chromic H. pylori infection is very common, with an assessed global mean prevalence of 58% (varies from 39.9% to 91.7%), partly due to immigrants coming from countries with a high prevalence of H. pylori infection [30,34,35]; it accounts of about 4.4 billion of the world’s H. pylori chronically infected individuals [36].
Despite the strong innate and adaptive immune responses, H. pylori exhibits long-term survival in the gastric mucosa. Its survival is intensely influenced by the ability of this bacterium to escape, disrupt, and manipulate the host immune responses. H. pylori employs several mechanisms to evade innate and adaptive immune systems’ immunity, allowing the bacterium to establish chronic infection [37]. H. pylori, by perturbing the gastric mucosa immune equilibrium, induces a microenvironment that favors gastric oncogenesis and evasion of immune surveillance [38]. This bacterium can escape from being recognized by innate immune receptors through changing its surface molecules. H. pylori can also block other innate recognition receptors by constraining downstream signaling pathways. Specifically, to the evasion of innate immune system mechanisms belong, among others, the following: neutralization of reactive oxygen and nitrogen species released by macrophages, alteration of cytokine secretion and the maturation of dendritic cells, and more passive systems such as the variability and the uniquely low immunogenicity of H. pylori lipopolysaccharide (LPS) [39]. Moreover, the expression of Lewis antigens in the LPS of H. pylori provides the bacterium with the ability to mimic host antigens and thus conceal itself from the immune system. H. pylori is also able to escape from the host adaptive immunity by modifying the function of the T lymphocytes. In this respect, H. pylori evasion from destruction by both the innate and adaptive immune responses may contribute to AD pathology [40].
It is important to note that diverse issues involved in the failure to eradicate H. pylori include, among others, H. pylori genotypes with high pathogenicity, the occurrence of point mutations, increased H. pylori burden, immunosuppressant characteristics, acidic gastric pH, intracellular occurrence of H. pylori, and/or biofilm creation linked with efflux pumps (transporters removing antibiotics from the cellular inner) [41]. H. pylori exhibits the ability to form biofilm in the stomach, which significantly decreases sensitivity to antibiotics and other antimicrobial agents and represents an independent mechanism that may be involved in the development and/or intensification of the antibiotic resistance. H. pylori biofilm appears to be a main reason of failure to eliminate infections triggered by H. pylori; antimicrobial agents are not capable of penetrating and destroying the biofilm; and H. pylori mutations resistant to antibiotics such as clarithromycin are commonly created in biofilms. Therefore, affecting the H. pylori biofilm-related pathologies can offer an efficient approach to avoid failures of H. pylori therapeutic regimens [41]. In this respect, exposure to key components of the innate immune system antimicrobial peptides, such as high human β-defensins (hβDs) 1–4 concentrations, significantly inhibits the capability for biofilm formation by H. pylori, a finding principally attributed to antibacterial action of hβDs [42]. Nevertheless, if H. pylori may access the brain, it could trigger the development and progression of neurodegenerative disorders, possibly by inducing defensin abnormal expression [43].
H. pylori also induces an intracellular niche that protects the bacterium from antibiotic eradication therapy, allowing persistence and recolonization. This intracellular niche may also contribute to bacterial evasion of the host immune responses [41]. In this regard, for instance, VDP, a direct vitamin D metabolite, possesses a H. pylori-specific antimicrobial ability through hβDs induction from infected macrophages, thus creating a promising therapeutic potential [41]. Notably, addition of vitamin D3 to the conventional clarithromycin-based triple therapeutic approach also offers a significant eradication rate of H. pylori infection.
Apart from defensins, another key component of the innate immune system, namely human cathelicidin LL-37, is able to prevent H. pylori biofilm creation by inhibiting H. pylori adhesion, downregulating biofilm-related genes, suppressing quorum-sensing pathways, degrading the biofilm matrix, and suppressing the biofilm-residing cells [44].
Autophagy is involved in H. pylori-induced gastritis, and chronic H. pylori infection promotes gastric malignancy growth and progress by considerably weakening autophagy; although autophagy offers an anti-carcinogenic effect in normal tissues, the occurrence of an excess of altering triggers, such as H. pylori infection, prevents its activity, thereby promoting cancer development [41]. Interestingly, circulating monocytes (infected with H. pylori due to defective autophagy) through a disrupted blood–brain barrier (BBB) may trigger neurodegeneration [45]. The clarification of the interaction among H. pylori and autophagic process has driven investigators to examine the effect of the regulators of the autophagic process in order to control H. pylori infection in an effort to improve the rising antibiotic resistance. However, future research is warranted to elucidate in depth the above-mentioned considerations.
Recurrence/reinfection of H. pylori infection can occur, and it can be categorized into two distinct mechanisms: recrudescence and reinfection [46]. The recrudescence is the reappearance of the original strain following initial eradication; it is considered to be a failure of eradication. The reinfection is infected with a new strain after initial eradication. The identification of the H. pylori strain needs the application of certain molecular fingerprinting techniques. These techniques are very complex procedures and demand high personnel and facilities resources. Therefore, currently, it cannot be widely introduced in clinical and scientific investigation. Some investigators consider that the recurrence of H. pylori infection less than 1 year after eradication is categorized as recrudescence but reinfection if it was more than 1 year [47,48]. Other investigators consider that the change of H pylori from negative to positive at 1 year following initial eradication might be caused by recurrence or reinfection, and reinfection is the principal cause of recurrence at 3 years after successful eradication [49]. A relative review and meta-analysis showed that the global annual recurrence, reinfection, and recrudescence rates of H. pylori were 4.3%, 3.1%, and 2.2%, respectively [50]. Risk factors of H. pylori reinfection include, among others, dental-plaque-related periodontitis, which is a multifactorial inflammatory disorder mediated by the host immune response to dental plaque [51,52]. Periodontitis is a risk factor for development of neurodegenerative disorders including AD, PD, or multiple sclerosis [53,54].
A large number of published studies cover H. pylori-preventing strategies, including dietary regimens. In general, dietary intervention to prevent H. pylori-related severe pathologies might be ideal because it carries no risk of bacterial resistance, safety, and rejuvenating action of atrophic gastritis. A few recent examples are the following:
Soy product consumption significantly reduced the incidence of gastric cancer [55]. This could be attributed to the isoflavones (e.g., genistein, daidzein, and glycitein) that are abundant in soy products. In particular, genistein inhibits the growth of H. pylori and the activity of nuclear factor (NF)-κB, which is associated with gastrointestinal tract tumorigenesis [55].
Cranberry supplementation appears to improve the success of eradication of H. pylori infection, which represents the major cause of peptic ulcer disease and gastric cancer [56].
Vitamins A, C, D, E, B6, B9, and B12 as well as key minerals such as zinc, iron, copper, nickel, and selenium may be involved in H. pylori infection and eradication. The antioxidant activity of vitamins can reduce oxidative stress and further suppress H. pylori-induced inflammation. Moreover, in vitro studies demonstrated promising anti-H. pylori mechanisms for vitamin administration in a dose-dependent manner. Antioxidants including certain vitamins may exhibit a preventative and therapeutic potential against AD, thereby offering insights into the association between oxidative stress and neurodegenerative disorders [57].
The administration of probiotics seems to reduce H. pylori adhesion to gastric epithelial cells and prevent H. pylori colonization, particularly in children, or reinfection with H. pylori in high-risk adult patients [58]. In this respect, recent data indicate that probiotic supplementation also exhibits a highly significant effect on cognitive function in patients with both cognitive impairment and AD [59].
Maintaining good hygiene is effective strategy in preventing H. pylori infection [60]. Likewise, maintaining good hygiene (oral) is an effective strategy in improving the clinical condition of patients with AD [61].
However, such H. pylori-preventive strategies warrant further evaluation.
Interestingly, H. pylori is also related to metabolic syndrome (MetS)-systemic pathologies such as cardio-cerebrovascular and neurodegenerative diseases, the end outcomes of MetS [22,62].
In this respect, by introducing gastric mucosa histology representing the practical diagnostic gold standard for active burden of H. pylori infection, this infection, as in the case of the patients with neurodegenerative disorders, is also connected with BCT even in cognitively normal people. This connection persists despite further adjustment for MetS risk factors, thereby indicating that the active burden of H. pylori infection may contribute to neurodegeneration in cognitively normal people [63]. In contrast, H. pylori eradication regimens may ameliorate and/or prevent both local and systemic chronic H. pylori infection-related disorders. For instance, in a topical setting, H. pylori eradication seems to reduce and/or prevent autoimmune gastritis as indicated by improvement of gastric atrophy on follow-up, mucosa-associated lymphoid tissue lymphoma, gastric adenocarcinoma, as well as metachronous gastric cancer and preneoplastic lesions including atrophic gastritis and intestinal metaplasia [64,65]. In a systemic setting, H. pylori eradication may exert a positive effect on AD manifestations at 5-year survival endpoints [63]. Likewise, eradication of this bacterium could constrain the progression of clinically isolated syndrome to the development of multiple sclerosis [25]. Moreover, H. pylori eradication results in significant clinical improvement in the symptoms and better PD patient outcomes [66,67,68]. It is important to note that abnormal BCT may contribute to cognitive decline in patients with PD [69,70]. Furthermore, regarding glaucoma (defined as “ocular” AD), we reported a 2-year beneficial effect of H. pylori eradication on glaucoma progression, suggesting a possible causal link between this bacterium and glaucoma [45,71]. BCT may also contribute to the severity and progression of glaucoma, thereby serving as a novel marker for assessing the severity of glaucoma [72,73].
Various H. pylori-related potential mechanisms that trigger neurodegeneration include the following: H. pylori-induced systemic inflammatory responses; H. pylori-induced metabolic dysfunctions; H. pylori-related direct brain injury by inducing toxic materials or by attacking the brain via gastrointestinal retrograde axonal transport or oral-nasal olfactory pathways; circulated H. pylori-infected monocytes that enter the brain due to disruption of BBB (Trojan horse concept) and induce neurodegeneration; and/or H. pylori-related gastrointestinal microbiota dysbiosis that modifies the gut–brain axis toward neurodegeneration [63] (Figure 1).
More specifically, H. pylori infection induces the release of a variety of molecules, such as IFN-γ, TNF-α, CRP, and IL-1, IL-6, and IL-10, all of which may promote apoptosis of neural tissue [74]. Additionally, structural components of H. pylori exhibit direct pro-inflammatory properties. For instance, H. pylori (2–20) peptides expressed by H. pylori, together with amyloid-β42, act as a ligand for formyl peptide receptors that play a key role in inflammation and lead to neurodegeneration. H. pylori (2–20)-mediated signaling results in increase of the vascular endothelial growth factor, a well-described mediator predisposing to vascular and other neurodegenerative dementias [75]. The direct pro-inflammatory properties of H. pylori infection components are also supported by experimental studies, according to which soluble surface fractions of H. pylori infection may lead to cognitive impairment by promoting the formation of Aβ42, thus compromising synaptic integrity and mediating neuronal damage [76]. Moreover, H. pylori infection associated with MetS may contribute to the vascular pathology, a well-described predisposing factor for neurodegenerative disease, by inducing increased plasma homocysteine levels and reduced serum 5-methyltetrahydrofolate and vitamin B-12, thus subsequently promoting endothelial damage. Interestingly, individuals with H. pylori infection exhibited cognitive impairment, the degree of which correlated with decreased 5-methyltetrahydrofolate levels [77]. H. pylori infection is also associated with increased tau-hyperphosphorylation, the key component of neurofibrillary tangles in AD [78]; it induces tau hyperphosphorylation, which is directly linked to the AD-associated neurodegeneration [79]. Intraperitoneal inoculation of H. pylori filtrate in rats provokes spatial learning and memory deficits, abnormal hippocampal dendritic spine maturation, and augmented presenilin-2 and Aβ42 in the brain’s hippocampus and cortex. Furthermore, H. pylori filtrate stimulates considerable tau hyperphosphorylation in mouse neuroblastoma N2a [76]. Since phosphorylation of tau is attenuated by the glycogen synthase kinase 3 inhibitor, this provides preliminary evidence for a beneficiary role of H. pylori eradication in AD and the need for verification by large-scale clinical trials [76]. Furthermore, we recently reviewed potential Galectin (Gal)-3-mediated mechanisms by which H. pylori may also promote neurodegeneration [27]. Gal-3 is a glycan-binding protein implicated in several physiologic and pathological processes including cell signaling, proliferation, and migration as well as the stimulation of immune response. H. pylori infection may promote neuroinflammation and neurodegeneration by several Gal-3-dependent pathways [27,80,81]. In addition, H. pylori-derived outer membrane vesicles modulate the physiological functions of glial cells and neurons and can worsen AD pathology [82]. H. pylori infection and/or gut microbiota alteration by promoting chronic inflammation, may influence both the occurrence and evolution of AD [83] (Figure 1). H. pylori, by releasing several inflammatory mediators (e.g., cytokines and chemokines induced by H. pylori infection), may induce BBB/blood–ocular barrier (BOB) breakdown, thereby being involved in the pathogenesis of neuropathies such as AD and glaucoma. TNF-α, induced by H. pylori infection, is involved in BBB disruption via a mechanism involving matrix metalloproteinase upregulation [62]. H. pylori VacA cytotoxin promotes intracellular survival of H. pylori, and activated monocytes (possibly infected with H. pylori due to defective autophagy) might access the brain (Trojan horse theory) via BBB/BOB disruption, thereby triggering the development and progression of neurodegenerative disorders, possibly by inducing additional defensin abnormal expression; when H. pylori accesses the brain, it may trigger defensin-related dendritic cells maturation and activation, leading to pro-inflammatory cytokine release by effector T cells, thereby promoting neuronal cell injury and death [24]. Furthermore, H. pylori could further contribute to neurodegenerative disorders’ pathogenesis by inducing MetS-related platelet–leukocyte aggregation, which is proposed to play pathophysiologic roles in AD; producing reactive oxygen species and circulating lipid peroxides implicated in AD pathobiology; and influencing the apoptotic process, a crucial cell death form in AD [66].
Therefore, controlling H. pylori infection may exhibit beneficial effects on neurodegeneration [22,62].
Additional critical triggers, particularly H. pylori-induced hyperhomocysteinemia, may play a crucial role in brain neurodegenerative disorders by provoking BCT in both normal males and females [84]. Since BCT, identified during the prodromal stages of AD, is a “candidate” biomarker implemented in AD clinical therapy trials, H. pylori eradication regimens, by affecting such BCT triggers, may exhibit a positive impact on cortical thickness, thereby signifying a relative protecting role in the development of brain-degenerative pathologies.
Recently, genetic polymorphisms in genes that have been implicated in AD, a primary neurodegenerative disease and the most common cause of dementia worldwide [85], include, among others, ABCA7 [86], PICALM [87], CLU [88], SLC24A4/RIN3 [89], ECHDC3 [90], MTHFR [91], as well as in insulin signaling such as INSR [92]. MTHFR mutations are associated with homocysteinemia, and increased homocysteine blood levels are considered to be an independent risk factor of cerebrovascular incidents. There are two common polymorphic genetic variants of the MTHFR gene that can lead to impaired functioning of the conversion of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate, a co-substrate for homocysteine remethylation to methionine, leading to hyperhomocysteinemia, which is a risk factor for dementia owing to an associated BBB dysfunction and subsequent small-vessel disease pathology [93]. Of note, the C677T polymorphism on the MTHFR gene can affect migraine susceptibility, with the resultant migraine being due to the increase in homocysteine levels in the blood [94]. Mutations in the gene SCIMP, a gene implicated in TLR-dependent activation of microglia, the innate immune system cell of the CNS, also have been recently identified in AD [95], thus shedding light onto possible mechanisms through which microbes and other infectious agents may affect the CNS in the context of neurodegeneration.
Specifically, atrophic gastritis due to H. pylori infection-related MetS provokes deficiency of both vitamin B12 and folic acid, leading to hyperhomocysteinemia with consequent injury of the vascular endothelium [96]; H. pylori-provoked chronic gastritis leads to defective absorption of vitamin B12 and folate, resulting in methylation by 5-methyl-tetrahydrofolic acid malfunction and, therefore, in hyperhomocysteinemia [96] (Figure 2). More specifically, homocysteine, a sulfur-containing amino acid derived from methionine, is principally metabolized via methionine-synthase in the remethylation cycle, which is dependent on the presence of both vitamin B12 and folate as co-factors. The mechanisms of vitamin B12 malabsorption by H. pylori infection that result in hyperhomocysteinemia are unclear, but the following explanations are possible: First, as acid and pepsin are critical for splitting vitamin B12 from food binders and for its subsequent transfer to R binder in the stomach, the diminished acid secretion in H. pylori-induced gastritis may lead to a failure in absorption of food-bound vitamin B12. Second, H. pylori-induced gastritis could cause a secretory dysfunction of the intrinsic factor, thus leading to malabsorption of vitamin B12 and hyperhomocysteinemia. In addition, chronic H. pylori infection has been shown to decrease secretion of ascorbic acid from the gastric mucosa and to increase gastric pH. Low ascorbic acid in gastric juice or high pH of gastric juice have been demonstrated to produce reduced dietary folate absorption, also leading to hyperhomocysteinemia [97]. Of note, plasma homocysteine concentrations can range from 5 to 15 μmol/L in normal people [98], and elevated homocysteine levels of 15 μmol/L or higher are considered as indicators of hyperhomocysteinemia [99].
Hyperhomocysteinemia, as an atherogenic mediator, appears to be a risk factor for cardio-cerebrovascular diseases and AD, the end-points of MetS [62,100] (Figure 2). Moreover, the three “non-conventional” coronary artery disease risk factors, including hyperhomocysteinemia, hyperfibrinogenemia, and high lipoprotein-a, may stimulate the existence of arteriosclerosis and sequelae such as neurodegeneration [101,102]. More specifically, hyperhomocysteinemia has been involved in the pathophysiology of neurodegenerative diseases including AD. As an indicator of AD, increased homocysteine levels raise Aβ concentrations and/or deposition, thereby contributing to cognitive deterioration [79]. Increased homocysteine levels, with a subsequence insufficiency of methionine and S-Adenosyl-L-methionine and high S-Adenosyl-L-homocysteine, are linked with the reduced methylation capability. Inhibition of methylation reactions worsens the Aβ and tau pathologies also seen in H. pylori-associated AD [79,103]. Hyperfibrinogenemia also appears to be a risk factor of AD. An interaction between fibrinogen and Aβ in brain tissue with the possible creation of fibrinogen-Aβ plaques may be connected to cognitive dysfunction. High fibrinogen serum levels are associated with BCT [104], and fibrinogen plus Aβ complex formation is a hallmark of AD. Moreover, lipoprotein-a has been documented as a crucial factor in creation of Aβ plaques and exacerbation of AD.
Cognitively normal older individuals with elevated plasma concentrations of Aβ exhibit lower cognitive performance and higher homocysteine concentrations and BCT compared with those with reduced Aβ concentrations. Especially, such individuals with elevated Aβ1-40 exhibit a thinner temporal lobe, poorer self-perception of daily memory, and hyperhomocysteinemia [84]. Thinning of the temporal lobe appears to start eleven years before onset of cognitive impairment; BCT is a very sensitive indicator of neurodegenerative alterations, appearing decades before onset of AD clinical symptoms. Additional data also suggest that hyperhomocysteinemia is significantly associated with BCT, indicating cerebral atrophy in elderly asymptomatic populations [105] (Figure 2). Our findings show that active H. pylori infection appears to contribute to the pathogenesis of mild cognitive impairment, a prodromic condition predisposing for subsequent development of AD, by triggering the following sequence: chronic atrophic gastritis - hyperhomocysteinemia - mild cognitive impairment - AD development [23]. Since mild cognitive impairment is significantly connected with BCT [106], further research is necessary to evaluate whether lowering plasma homocysteine concentrations in both asymptomatic people and mild cognitive impairment patients with H. pylori infection may prevent neurodegenerative changes or delay the progression to the development of dementia.
Specifically, hyperhomocysteinemia appears to play an active role in the pathophysiology of mild cognitive impairment – AD sequence by triggering Aβ and tau pathologic conditions linked with synaptic dysfunction, neuroinflammatory process, and memory worsening (Figure 2). This finding signifies a potential novel strategy for the therapy of populations exhibiting this risk parameter [107]. In this respect, apart from increased levels of homocysteine, other H. pylori-associated MetS conditions provoke the mentioned mild cognitive impairment – AD sequence [62]. Experimental evidence indicates that only live H. pylori filtrates injection and not Escherichia coli induce in rats spatial learning and memory insufficiency [76]. Moreover, live H. pylori filtrates raise Aβ42 in the hippocampus and cortex by augmenting γ-secretase activity, thus triggering cognitive deterioration via interruption of the synaptic function [76] (Figure 2). Likewise, H. pylori filtrates stimulate tau hyperphosphorylation in rats at many AD-linked tau phosphorylation brain areas with activation of glycogen synthase kinase-3β (GSK-3β) [76] (Figure 2).
In contrast, GSK-3 inhibitors seem to attenuate the H. pylori-provoked tau hyperphosphorylation. These data provide an indication for potential involvement of H. pylori infection in AD comparable tau pathology and suggest that H. pylori eradication might exert a beneficial effect against tauopathy [76]. The aforementioned experimental data seem to support our own clinical studies showing that H. pylori eradication may exert a positive effect on AD manifestations at 2- and 5-year survival endpoints [108]. Persistent H. pylori infection above 3 years results in hyperhomocysteinemia in healthy people [109]. Likewise, H. pylori eradication in a vitamin-B12-deficiency setting leads to increased vitamin B12 levels and reduced homocysteine blood levels [110]. Therefore, additional studies are needed to clarify the influence of eradication of this bacterium in the following sequence: hyperhomocysteinemia-linked asymptomatic setting - mild cognitive impairment - AD.
PD, a major worldwide health problem also associated with AD [111,112], is a neurodegenerative disease of the CNS affecting primarily dopaminergic neurons in the substantia nigra.
Alpha-synuclein (α-synuclein) is a protein participating in the transport of synaptic vesicles and the release of neurotransmitters in the synaptic cleft. Moreover, the α-synuclein protein is crucially involved in PD pathology, as the aggregation of α-synuclein is associated with neuron loss in PD. More specifically, α-synuclein misfolding leads to toxicity mediated by a-synuclein aggregates, namely protofibrils, that cause loss of synapses and neuronal death (Figure 1). In this respect, α-synuclein-associated mutations, for instance, in the SNCA gene, are related not only with familial PD but also predispose to sporadic disease. Pathological mechanisms that also contribute in PD evolution are mitochondrial dysfunction, the production of reactive oxygen species and other oxidative species, defective calcium homeostasis, endoplasmic reticulum/Golgi impairment, and microglia activation in the context of neuroinflammation [113]. Apart from the motor dysfunction in PD, non-motor symptoms have been the focus of extensive research, such as impairment of olfaction, mood disorders, and cognitive deficit [114,115]. Advanced neuroimaging biomarkers have been increasingly under study in the context of PD, and their association with cognitive function and other non-motor symptoms is under thorough investigation. More specifically, in PD, BCT has been associated with mild cognitive impairment [116] and rapid eye movement sleep behavior disorder [117], whereas it has been suggested as a possible biomarker with prognostic potential [118,119]. Changes in cortical thickness may lead to a dysfunction of resting-state functional connectivity, contributing to cognitive decline in patients with PD [69].
Recent meta-analysis indicates that both PD and AD are significantly linked with gastrointestinal disorders including H. pylori infection. Therefore, the adverse role of H. pylori in the pathophysiology of PD or AD should be evaluated to shed new light on the diagnosis and therapy of PD and AD [120].
Focusing on H. pylori-related PD, relative data indicate that H. pylori infection may play a role in PD pathophysiology. There is a strong association between H. pylori chronic infection-related gastric pathologies and exacerbation of PD symptoms [121]. A large cross-sectional study demonstrated a positive connection among H. pylori positivity (detected by 13C-urea breath test) and PD motor severity [121]. Likewise, H. pylori infection and MetS are important risk factors for PD [66]. Apoptotic microglia-associated nerve cell death seems to underlie PD and other common neurological conditions including AD, multiple sclerosis, and glaucoma, which are also associated with H. pylori infection [66].
In this regard, hyperhomocysteinemia could drive PD development and progression via many pathways involving apoptosis, oxidative stress, mitochondrial dysfunction, and DNA damage in nerve cells [122]. In PD, hyperhomocysteinemia seems to be connected with cognitive performance and structural damage in the cerebral cortex [123]. Hyperhomocysteinemia is a risk factor for cognitive decline in PD [124]. Thus, hyperhomocysteinemia during PD development and progression offers a novel strategy for the diagnosis and treatment of PD [122]. Relative data indicate the occurrence of hyperhomocysteinemia in levodopa-treated PD patients, whereas vitamin B12 and folate supplementation may decrease homocysteine levels [125]. Likewise, H. pylori eradication in patients with PD might improve the bioavailability of L-3,4-dihydroxyphenylalanine (L-dopa), a precursor of dopamine used as a treatment for PD, and reduce motor fluctuations [111]. In addition, H. pylori, which selectively infects and colonizes predominantly the gastric epithelium and oral cavity [126], is a risk factor for periodontitis [127]; periodontitis, beyond AD [53], is linked with PD [128,129]. In contrast, H. pylori eradication and treatment of periodontitis are beneficial for the prevention of PD and dementia [127].
Equivalent data may also be obtained for clinically isolated syndrome - multiple sclerosis sequence [25,28,130]. Indeed, our studies show that active H. pylori infection, apart from multiple sclerosis [28,130], is also common in clinically isolated syndrome, a prodromic condition predisposing for subsequent development of multiple sclerosis, accompanied by increased homocysteine levels. Moreover, eradication of this bacterium could constrain the progression of clinically isolated syndrome to the development of multiple sclerosis [25]; BCT is observed in both clinically isolated syndrome and multiple sclerosis [130]. In addition, recent data indicate that hyperhomocysteinemia, beyond cardiovascular disorders, appears to be an important factor involved in the pathogenesis of such an autoimmune disease of the CNS system as multiple sclerosis; it plays a role in endothelial dysfunction in multiple sclerosis [131]. Hyperhomocysteinemia is linked with high process activity and disease progression by involving in the mechanisms of neurodegeneration observed in multiple sclerosis. Thus, homocysteine concentration may be a potential marker predicting the course of the disease [132]. Another study reported higher homocysteine blood levels in patients with progressive multiple sclerosis compared to patients with relapsing-remitting multiple sclerosis [133]. Of note, progressive multiple sclerosis is a form of the disease typically considered to exhibit higher neurodegenerative components in terms of underlying pathology, whereas relapsing-remitting multiple sclerosis is primarily characterized by increased relative contribution of neuroinflammation in the disease pathology [134]. In this respect, the findings of the study underline a possible mechanism by which H. pylori infection and H. pylori infection-related hyperhomocysteinemia may contribute to neurodegeneration in the context of multiple sclerosis.
Glaucoma, the leading cause of irreversible blindness worldwide, is linked with both PD and AD [135].
Specifically, active H. pylori infection, beyond PD, is also associated with primary open-angle glaucoma, which has been called “ocular AD”, and pseudo-exfoliative glaucoma in some populations studied [136,137,138]. Moreover, H. pylori eradication seems to improve glaucoma parameters, thereby signifying a potential causal relationship between H. pylori infection and glaucoma [136]. Glaucoma induces the death of retinal ganglion cells and their axons and appears to be the second most common reason of blindness worldwide [138]. Although glaucoma is frequently considered an eye-only disorder, relative data suggest the involvement of the brain’s visual system in glaucoma. Post-mortem histology has disclosed that signs of neurodegeneration and associated metabolic changes are not restricted to retinal ganglion cells and the optic nerve but encompass upstream parts of the central visual pathway including the visual cortex. According to the mentioned BCT analyses, cortical degeneration within the visual areas becomes progressively aggravated with the augmenting severity of primary open-angle glaucoma [139]. The cortical thinning of visual areas might play a key role in the progression of glaucoma and can be a novel biomarker for assessing the severity of glaucoma [139]. Moreover, the inner retinal layer thinning, optic nerve cupping, and reduced visual cortex activity may occur before patients show visual field impairment [72].
In this respect, hyperhomocysteinemia may play a role in the pathogenesis of retinal diseases, especially glaucoma and age-related macular degeneration [140]. Oxidative stress and mitochondrial dysfunction are main mediators involved in the pathogenesis of neurodegenerative and in inner retinal neurodegenerative disorders. These mediators are also mediators of hyperhomocysteinemia-induced neuronal retinal ganglion cells death [141]; and oxidative stress is a major pathogenic mechanism related to hyperhomocysteinemia. Particularly, as in the case of H. pylori infection [137,138,142], hyperhomocysteinemia induces endothelial dysfunction and is associated with primary open-angle glaucoma and/or pseudo-exfoliation glaucoma, thereby signifying the potential prevention and therapy of glaucoma by regulating homocysteine levels [143]. Moreover, nutritional disturbances seem to be connected with an augmented risk of glaucoma [139], and the treatment of hyperhomocysteinemia by receiving low-cost vitamin B12 and folic acid regimens may prevent vascular abnormalities of this disorder [144]. Of note, early and long-term vitamin B12 supplementation may offer benefits against cognitive decline [145]. Nevertheless, whether H. pylori eradication, beyond other glaucoma parameters, offers an additional benefit regarding hyperhomocysteinemia-associated glaucoma remains to be elucidated in the future.
Finally, BCT in schizophrenia might be correlated with subsequent suicidal behavior [146]. In this regard, hyperhomocysteinemia appears to play a role in the pathophysiology of schizophrenia; hyperhomocysteinemia is correlated with negative symptoms of schizophrenia [147], and it may serve as a prognostic marker for schizophrenia [148]. Likewise, H. pylori has been proposed to play a role as a potential environmental factor for pathogenesis of schizophrenia [149], and H. pylori infection, associated with homocysteine changes, might contribute to schizophrenia development [150]. Moreover, both schizophrenia and H. pylori infection have been correlated with malnutrition [151,152], and H. pylori infection, among others, induces the reduced bioavailability or the malabsorption of essential nutrients such as the mentioned vitamin B12 and folate, leading to hyperhomocysteinemia [151]. Vitamin supplementation, especially with low-cost vitamin B12, folate, and vitamin D, could offer significant benefits in the treatment of schizophrenia [153] and could improve the H. pylori eradication rate [154]. Nevertheless, whether H. pylori eradication may also offer benefit in the treatment of schizophrenia requires further investigation.
In conclusion, controlling the impact of H. pylori-related hyperhomocysteinemia on such neurodegenerative disorders might offer benefits, and additional research is warranted to clarify this crucial topic currently representing a major global burden.

Author Contributions

J.K. conceptualized the idea, developed the original draft, and finalized the manuscript; M.D., A.P., S.A.P., C.Z., E.K., S.A., F.K., M.C.M., M.B. and E.V. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable for opinion article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yadav, P.; Lee, Y.-H.; Panday, H.; Kant, S.; Bajwa, N.; Parashar, R.; Jha, S.K.; Jha, N.K.; Nand, P.; Lee, S.S.; et al. Implications of Microorganisms in Alzheimer’s Disease. Curr. Issues Mol. Biol. 2022, 44, 4584–4615. [Google Scholar] [CrossRef] [PubMed]
  2. Doulberis, M.; Kotronis, G.; Gialamprinou, D.; Polyzos, S.A.; Papaefthymiou, A.; Katsinelos, P.; Kountouras, J. Alzheimer’s disease and gastrointestinal microbiota; impact of Helicobacter pylori infection involvement. Int. J. Neurosci. 2021, 131, 289–301. [Google Scholar] [CrossRef] [PubMed]
  3. Sait, A.; Angeli, C.; Doig, A.J.; Day, P.J.R. Viral Involvement in Alzheimer’s Disease. ACS Chem. Neurosci. 2021, 12, 1049–1060. [Google Scholar] [CrossRef] [PubMed]
  4. Alegret, M.; Vinyes-Junqué, G.; Boada, M.; Martínez-Lage, P.; Cuberas, G.; Espinosa, A.; Roca, I.; Hernàndez, I.; Valero, S.; Rosende-Roca, M.; et al. Brain Perfusion Correlates of Visuoperceptual Deficits in Mild Cognitive Impairment and Mild Alzheimer’s Disease. J. Alzheimer’s Dis. 2010, 21, 557–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Fjell, A.M.; Walhovd, K.B. Structural Brain Changes in Aging: Courses, Causes and Cognitive Consequences. Rev. Neurosci. 2010, 21, 187–221. [Google Scholar] [CrossRef]
  6. Gupta, Y.; Lee, K.H.; Choi, K.Y.; Lee, J.J.; Kim, B.C.; Kwon, G.-R. Alzheimer’s Disease Diagnosis Based on Cortical and Subcortical Features. J. Healthc. Eng. 2019, 2019, 2492719. [Google Scholar] [CrossRef]
  7. Zarei, M.; Ibarretxe-Bilbao, N.; Compta, Y.; Hough, M.; Junque, C.; Bargallo, N.; Tolosa, E.; Jose Martí, M. Cortical thinning is associated with disease stages and dementia in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 2013, 84, 875–882. [Google Scholar] [CrossRef] [Green Version]
  8. Chen, Q.; Hattori, T.; Tomisato, H.; Ohara, M.; Hirata, K.; Yokota, T. Turning and multitask gait unmask gait disturbance in mild-to-moderate multiple sclerosis: Underlying specific cortical thinning and connecting fibers damage. Hum. Brain Mapp. 2023, 44, 1193–1208. [Google Scholar] [CrossRef]
  9. Zhao, Y.; Zhang, Q.; Shah, C.; Li, Q.; Sweeney, J.A.; Li, F.; Gong, Q. Cortical Thickness Abnormalities at Different Stages of the Illness Course in Schizophrenia. JAMA Psychiatry 2022, 79, 560. [Google Scholar] [CrossRef]
  10. Cheng, C.P.-W.; Cheng, S.-T.; Tam, C.W.-C.; Chan, W.-C.; Chu, W.C.-W.; Lam, L.C.-W. Relationship between Cortical Thickness and Neuropsychological Performance in Normal Older Adults and Those with Mild Cognitive Impairment. Aging Dis. 2018, 9, 1020. [Google Scholar] [CrossRef] [Green Version]
  11. Machulda, M.M.; Lundt, E.S.; Albertson, S.M.; Spychalla, A.J.; Schwarz, C.G.; Mielke, M.M.; Jack, C.R.; Kremers, W.K.; Vemuri, P.; Knopman, D.S.; et al. Cortical atrophy patterns of incident MCI subtypes in the Mayo Clinic Study of Aging. Alzheimer’s Dement. 2020, 16, 1013–1022. [Google Scholar] [CrossRef]
  12. Hayes, J.P.; Pierce, M.E.; Brown, E.; Salat, D.; Logue, M.W.; Constantinescu, J.; Valerio, K.; Miller, M.W.; Sherva, R.; Huber, B.R.; et al. Genetic Risk for Alzheimer Disease and Plasma Tau Are Associated With Accelerated Parietal Cortex Thickness Change in Middle-Aged Adults. Neurol. Genet. 2023, 9, e200053. [Google Scholar] [CrossRef]
  13. Sampedro, F.; Puig-Davi, A.; Martinez-Horta, S.; Pagonabarraga, J.; Horta-Barba, A.; Aracil-Bolaños, I.; Kulisevsky, J. Cortical macro and microstructural correlates of cognitive and neuropsychiatric symptoms in Parkinson’s disease. Clin. Neurol. Neurosurg. 2023, 224, 107531. [Google Scholar] [CrossRef]
  14. Yang, D.; Masurkar, A.V.; Khasiyev, F.; Rundek, T.; Wright, C.B.; Elkind, M.S.V.; Sacco, R.L.; Gutierrez, J. Intracranial artery stenosis is associated with cortical thinning in stroke-free individuals of two longitudinal cohorts. J. Neurol. Sci. 2023, 444, 120533. [Google Scholar] [CrossRef]
  15. Dounavi, M.-E.; Newton, C.; Jenkins, N.; Mak, E.; Low, A.; Muniz-Terrera, G.; Williams, G.B.; Lawlor, B.; Naci, L.; Malhotra, R.; et al. Macrostructural brain alterations at midlife are connected to cardiovascular and not inherited risk of future dementia: The PREVENT-Dementia study. J. Neurol. 2022, 269, 4299–4309. [Google Scholar] [CrossRef]
  16. Shaw, M.E.; Abhayaratna, W.P.; Sachdev, P.S.; Anstey, K.J.; Cherbuin, N. Cortical Thinning at Midlife: The PATH Through Life Study. Brain Topogr. 2016, 29, 875–884. [Google Scholar] [CrossRef]
  17. Vidal-Piñeiro, D.; Sørensen, Ø.; Blennow, K.; Capogna, E.; Halaas, N.B.; Idland, A.-V.; Mowinclkel, A.M.; Pereira, J.B.; Watne, L.O.; Zwttweberg, H.; et al. Relationship between cerebrospinal fluid neurodegeneration biomarkers and temporal brain atrophy in cognitively healthy older adults. Neurobiol. Aging 2022, 116, 80–91. [Google Scholar] [CrossRef]
  18. Itzhaki, R.F. Herpes and Alzheimer’s Disease: Subversion in the Central Nervous System and How It Might Be Halted. J. Alzheimer’s Dis. 2016, 54, 1273–1281. [Google Scholar] [CrossRef]
  19. Frölich, L. Alzheimer’s disease—The ‘microbial hypothesis’ from a clinical and neuroimaging perspective. Psychiatry Res. Neuroimaging 2020, 306, 111181. [Google Scholar] [CrossRef]
  20. Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Grigoriadis, N.; Tsolaki, M.; Chatzopoulos, D.; Katsinelos, P.; Tzilves, D.; et al. Increased Cerebrospinal Fluid Helicobacter pylori Antibody in Alzheimer’s Disease. Int. J. Neurosci. 2009, 119, 765–777. [Google Scholar] [CrossRef]
  21. Kountouras, J.; Tsolaki, M.; Gavalas, E.; Boziki, M.; Zavos, C.; Karatzoglou, P.; Chatopoulow, D.; Venizelos, I. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology 2006, 66, 938–940. [Google Scholar] [CrossRef] [PubMed]
  22. Kountouras, J.; Papaefthymiou, A.; Polyzos, S.A.; Liatsos, C.; Tzitiridou-Chatzopoulou, M.; Chatzopoulos, D.; Vardaka, E.; Gilambrinou, D.; Kotronis, G.; Doulberis, M. Potential impact of Helicobacter pylori and metabolic syndrome-related non-alcoholic fatty liver disease on cardio-cerebrovascular disease. Metabolism 2022, 135, 155276. [Google Scholar] [CrossRef] [PubMed]
  23. Kountouras, J.; Tsolaki, M.; Boziki, M.; Gavalas, E.; Zavos, C.; Stergiopoulos, C.; Kapetanakis, N.; Chatzopoulos, D.; Venizelos, I. Association between Helicobacter pylori infection and mild cognitive impairment. Eur. J. Neurol. 2007, 14, 976–982. [Google Scholar] [CrossRef] [PubMed]
  24. Kountouras, J.; Zavos, C.; Polyzos, S.A.; Deretzi, G. Potential impact of Helicobacter pylori-related human β-defensin-1 on hepatic encephalopathy and neurodegeneration. Ann. Gastroenterol. 2016, 29, 99. [Google Scholar]
  25. Deretzi, G.; Gavalas, E.; Boziki, M.; Tsiptsios, D.; Polyzos, S.A.; Venizelos, I.; Zavos, C.; Koutlas, E.; Tsiptsios, I.; Katsinelos, P.; et al. Impact of Helicobacter pylori on multiple sclerosis-related clinically isolated syndrome. Acta Neurol. Scand. 2016, 133, 268–275. [Google Scholar] [CrossRef]
  26. Kountouras, J.; Boziki, M.; Polyzos, S.A.; Katsinelos, P.; Gavalas, E.; Zeglinas, C.; Tzivras, D.; Romiopoulos, I.; Giorgakis, N.; Anastasiadou, K.; et al. Impact of reactive oxygen species generation on Helicobacter pylori-related extragastric diseases: A hypothesis. Free Radic. Res. 2017, 51, 73–79. [Google Scholar] [CrossRef]
  27. Boziki, M.; Polyzos, S.A.; Deretzi, G.; Kazakos, E.; Katsinelos, P.; Doulberis, M.; Kotronis, G.; Giartza-Taxidou, E.; Laskaridis, L.; Tzivras, D.; et al. A potential impact of Helicobacter pylori -related galectin-3 in neurodegeneration. Neurochem. Int. 2018, 113, 137–151. [Google Scholar] [CrossRef]
  28. Arjmandi, D.; Abdollahi, A.; Ardekani, A.; Razavian, I.; Razavian, E.; Sartip, B.; Mahjour, S.; Parsa, H.; Kyvanasi, N.A.; Marhoommirzabak, E.; et al. Helicobacter pylori infection and risk of multiple sclerosis: An updated meta-analysis. Helicobacter 2022, 27, e12927. [Google Scholar] [CrossRef]
  29. Sankararaman, S.; Moosavi, L. Urea Breath Test. Book. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 January, 8 August 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK542286/ (accessed on 23 December 2022).
  30. Kountouras, J.; Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A.; Vardaka, E.; Tzivras, D.; Dardiotis, E.; Deretzi, G.; Giartza-Taxidou, E.; Grigoriadis, S.; et al. A perspective on risk factors for esophageal adenocarcinoma: Emphasis on Helicobacter pylori infection. Ann. N. Y. Acad. Sci. USA 2019, 1452, 12–17. [Google Scholar] [CrossRef]
  31. Kountouras, J.; Papaefthymiou, A.; Doulberis, M.; Polyzos, S.A. Influence of Helicobacter pylori -connected metabolic syndrome on non-alcoholic fatty liver disease and its related colorectal neoplasm high risk. Liver Int. 2020, 40, 475–476. [Google Scholar] [CrossRef]
  32. Kountouras, J.; Polyzos, S.A.; Doulberis, M.; Zeglinas, C.; Artemaki, F.; Vardaka, E.; Deretzi, G.; Giartza-Taxidou, E.; Tsivras, D.; Vlachaki, E.; et al. Potential impact of Helicobacter pylori-related metabolic syndrome on upper and lower gastrointestinal tract oncogenesis. Metabolism 2018, 87, 18–24. [Google Scholar] [CrossRef]
  33. Fagan-Garcia, K.; Geary, J.; Chang, H.-J.; McAlpine, L.; Walker, E.; Colquhoun, A.; van Zanten, S.V.; Girgis, S.; Archie, B.; Hanley, B.; et al. Burden of disease from Helicobacter pylori infection in western Canadian Arctic communities. BMC Public Health 2019, 19, 730. [Google Scholar] [CrossRef] [Green Version]
  34. Kountouras, J.; Papaefthymiou, A.; Polyzos, S.A.; Deretzi, G.; Vardaka, E.; Soteriades, E.S.; Tzitiridou-Chatzopoulou, M.; Gkolfakis, P.; Karafyllidou, K.; Doulbris, M. Impact of Helicobacter pylori-Related Metabolic Syndrome Parameters on Arterial Hypertension. Microorganisms 2021, 9, 2351. [Google Scholar] [CrossRef]
  35. Gerges, S.E.; Alosh, T.K.; Khalil, S.H.; el Din, M.M.W. Relevance of Helicobacter pylori infection in Egyptian multiple sclerosis patients. Egypt J. Neurol. Psychiatr. Neurosurg. 2018, 54, 41. [Google Scholar] [CrossRef] [Green Version]
  36. 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.; 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] [Green Version]
  37. Sijmons, D.; Guy, A.J.; Walduck, A.K.; Ramsland, P.A. Helicobacter pylori and the Role of Lipopolysaccharide Variation in Innate Immune Evasion. Front. Immunol. 2022, 13, 868225. [Google Scholar] [CrossRef]
  38. Olnes, M.J.; Martinson, H.A. Recent advances in immune therapies for gastric cancer. Cancer Gene Ther. 2021, 28, 924–934. [Google Scholar] [CrossRef]
  39. Lina, T.T. Immune evasion strategies used by Helicobacter pylori. World J. Gastroenterol. 2014, 20, 12753. [Google Scholar] [CrossRef]
  40. Miklossy, J.; McGeer, P.L. Common mechanisms involved in Alzheimer’s disease and type 2 diabetes: A key role of chronic bacterial infection and inflammation. Aging 2016, 8, 575–588. [Google Scholar] [CrossRef] [Green Version]
  41. Kountouras, J. Concentrating on the Long Topic of Empirical or Not Conventional Helicobacter pylori Eradication Regimens. Clin. Gastroenterol. Hepatol. 2022, S1542-3565(22)01016-3. [Google Scholar] [CrossRef]
  42. Kazakos, E.I.; Dorrell, N.; Polyzos, S.A.; Deretzi, G.; Kountouras, J. Comment on “Effect of biofilm formation by clinical isolates of Helicobacter pylori on the efflux-mediated resistance to commonly used antibiotics”. World J. Gastroenterol. 2017, 23, 6194–6196. [Google Scholar] [CrossRef] [PubMed]
  43. Kountouras, J.; Deretzi, G.; Gavalas, E.; Zavos, C.; Polyzos, S.A.; Kazakos, E. A proposed role of human defensins in Helicobacter pylori-related neurodegenerative disorders. Med. Hypotheses 2014, 82, 368–373. [Google Scholar] [CrossRef] [PubMed]
  44. Memariani, H.; Memariani, M. Antibiofilm properties of cathelicidin LL-37: An in-depth review. World J. Microbiol. Biotechnol. 2023, 39, 99. [Google Scholar] [CrossRef] [PubMed]
  45. Doulberis, M.; Kotronis, G.; Thomann, R.; Polyzos, S.A.; Boziki, M.; Gialamprinou, D.; Deretzi, G.; Katsinelos, P.; Kountouras, J. Impact of Helicobacter pylori on Alzheimer’s disease: What do we know so far? Helicobacter 2018, 23, e12454. [Google Scholar] [CrossRef]
  46. Sjomina, O.; Pavlova, J.; Niv, Y.; Leja, M. Epidemiology of Helicobacter pylori infection. Helicobacter 2018, 23, e12514. [Google Scholar] [CrossRef] [Green Version]
  47. Zhou, G. Helicobacter pylori Recurrence after Eradication Therapy in Jiangjin District, Chongqing, China. Gastroenterol. Res. Pract. 2020, 2020, 7510872. [Google Scholar] [CrossRef] [Green Version]
  48. Khor, C.J.; Fock, K.M.; Ng, T.M.; Teo, E.K.; Sim, C.S.; Tan, A.L.; Ng, A. Recurrence of Helicobacter pylori infection and duodenal ulcer relapse, following successful eradication in an urban east Asian population. Singap. Med. J. 2000, 41, 382–386. [Google Scholar]
  49. Xue, Y.; Zhou, L.Y.; Lu, H.P.; Liu, J.Z. Recurrence of Helicobacter pylori infection. Chin. Med. J. 2019, 132, 765–771. [Google Scholar] [CrossRef]
  50. Hu, Y.; Wan, J.H.; Li, X.Y.; Zhu, Y.; Graham, D.Y.; Lu, N.H. Systematic review with meta-analysis: The global recurrence rate of Helicobacter pylori. Aliment. Pharmacol. Ther. 2017, 46, 773–779. [Google Scholar] [CrossRef] [Green Version]
  51. Kini, V.; Mohanty, I.; Telang, G.; Vyas, N. Immunopathogenesis and distinct role of Th17 in periodontitis: A review. J. Oral Biosci. 2022, 64, 193–201. [Google Scholar] [CrossRef]
  52. Xie, Y.; Song, C.; Cheng, H.; Xu, C.; Zhang, Z.; Wang, J.; Huo, L.; DU, Q.; Xu, J.; Chen, Y.; et al. Long-term follow-up of Helicobacter pylori reinfection and its risk factors after initial eradication: A large-scale multicentre, prospective open cohort, observational study. Emerg. Microbes Infect. 2020, 9, 548–557. [Google Scholar] [CrossRef] [Green Version]
  53. Alvarenga, M.O.P.; Frazão, D.R.; de Matos, I.G.; Bittencourt, L.O.; Fagundes, N.C.F.; Rösing, C.K.; Maia, L.C.; Lima, R.R. Is There Any Association Between Neurodegenerative Diseases and Periodontitis? A Systematic Review. Front. Aging Neurosci. 2021, 24. [Google Scholar] [CrossRef]
  54. Li, X.; Kiprowska, M.; Kansara, T.; Kansara, P.; Li, P. Neuroinflammation: A Distal Consequence of Periodontitis. J. Dent. Res. 2022, 101, 1441–1449. [Google Scholar] [CrossRef]
  55. Siriviriyakul, P.; Werawatganon, D.; Phetnoo, N.; Somanawat, K.; Chatsuwan, T.; Klaikeaw, N. Chay Genistein attenuated gastric inflammation and apoptosis in Helicobacter pylori-induced gastropathy in rats. BMC Gastroenterol. 2020, 20, 410. [Google Scholar] [CrossRef]
  56. Piscione, M.; Mazzone, M.; di Marcantonio, M.C.; Muraro, R.; Mincione, G. Eradication of Helicobacter pylori and Gastric Cancer: A Controversial Relationship. Front. Microbiol. 2021, 12, 630852. [Google Scholar] [CrossRef]
  57. Varesi, A.; Campagnoli, L.I.M.; Carrara, A.; Pola, I.; Floris, E.; Ricevuti, G.; Chirumbolo, S.; Pascale, A. Non-Enzymatic Antioxidants against Alzheimer’s Disease: Prevention, Diagnosis and Therapy. Antioxidants 2023, 12, 180. [Google Scholar] [CrossRef]
  58. Marginean, C.M.; Cioboata, R.; Olteanu, M.; Vasile, C.M.; Popescu, M.; Popescu, A.I.S.; Bondari, S.; Pirscoveanu, D.; Marginean, I.C.; Iacob, G.A.; et al. The Importance of Accurate Early Diagnosis and Eradication in Helicobacter pylori Infection: Pictorial Summary Review in Children and Adults. Antibiotics 2022, 12, 60. [Google Scholar] [CrossRef]
  59. Liu, N.; Yang, D.; Sun, J.; Li, Y. Probiotic supplements are effective in people with cognitive impairment: A meta-analysis of randomized controlled trials. Nutr. Rev. 2023, nuac113. [Google Scholar] [CrossRef]
  60. Zhou, M.; Zeng, Y.; Xi, Y.; Luo, S.; Qi, J.; Zhao, G.; Guo, Y.; Cheng, F. School-based Hygiene Intervention to Prevent Helicobacter Pylori infection among childrEn (SHIP HOPE): Protocol for a cluster-randomised controlled trial. BMJ Open 2022, 12, e064207. [Google Scholar] [CrossRef]
  61. Hernández-Vásquez, A.; Barrenechea-Pulache, A.; Aguirre-Ipenza, R.; Comandé, D.; Azañedo, D. Interventions to Improve the Oral Hygiene of Individuals with Alzheimer’s Disease: A Systematic Review. Dent. J. 2022, 10, 92. [Google Scholar] [CrossRef]
  62. Kountouras, J.; Doulberis, M.; Polyzos, S.A.; Katsinelos, T.; Vardaka, E.; Kountouras, C.; Arapoglou, S.; Exadaktylos, A.; Deretzi, G.; Tsolaki, M.; et al. Impact of Helicobacter pylori and/or Helicobacter pylori-related metabolic syndrome on incidence of all-cause and Alzheimer’s dementia. Alzheimer’s Dement. 2019, 15, 723–725. [Google Scholar] [CrossRef] [PubMed]
  63. Park, J.; Kim, T.J.; Song, J.H.; Jang, H.; Kim, J.S.; Kang, S.H.; Kim, H.R.; Hwangbo, S.; Shin, H.Y.; Na, D.L.; et al. Helicobacter Pylori Infection Is Associated with Neurodegeneration in Cognitively Normal Men. J. Alzheimer’s Dis. 2021, 82, 1591–1599. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, J.; Wang, T.C. Helicobacter pylori and Gastric Cancer. Gastrointest. Endosc. Clin. 2021, 31, 451–465. [Google Scholar] [CrossRef] [PubMed]
  65. Kotera, T.; Nishimi, Y.; Kushima, R.; Haruma, K. Regression of Autoimmune Gastritis after Eradication of Helicobacter pylori. Case Rep. Gastroenterol. 2023, 17, 34–40. [Google Scholar] [CrossRef] [PubMed]
  66. Kountouras, J.; Boziki, M.; Polyzos, S.A.; Katsinelos, P.; Gavalas, E.; Zeglinas, C.; Tzivras, D.; Romiopoulos, I.; Giorgakis, N.; Anastasiadou, K.; et al. The Emerging Role of Helicobacter Pylori-Induced Metabolic Gastrointestinal Dysmotility and Neurodegeneration. Curr. Mol. Med. 2017, 17, 389–404. [Google Scholar] [CrossRef]
  67. Dardiotis, E.; Tsouris, Z.; Mentis, A.-F.A.; Siokas, V.; Michalopoulou, A.; Sokratous, M.; Dastamani, M.; Bogdanos, D.P.; Deretzi, G.; Kountouras, J. H. pylori and Parkinson’s disease: Meta-analyses including clinical severity. Clin. Neurol. Neurosurg. 2018, 175, 16–24. [Google Scholar] [CrossRef]
  68. Lolekha, P.; Sriphanom, T.; Vilaichone, R.-K. Helicobacter pylori eradication improves motor fluctuations in advanced Parkinson’s disease patients: A prospective cohort study (HP-PD trial). PLoS ONE 2021, 16, e0251042. [Google Scholar] [CrossRef]
  69. Zhu, Y.; Yang, B.; Zhou, C.; Gao, C.; Hu, Y.; Yin, W.F.; Yin, K.; Zhu, Y.; Jiang, G.; Ren, H.; et al. Cortical atrophy is associated with cognitive impairment in Parkinson’s disease: A combined analysis of cortical thickness and functional connectivity. Brain Imaging Behav. 2022, 16, 2586–2600. [Google Scholar] [CrossRef]
  70. Gu, L.; Shu, H.; Wang, Y.; Xu, H. Exploring brain changes of impulse control disorders in Parkinson’s disease: An ALE study. Front. Aging Neurosci. 2022, 14, 966525. [Google Scholar] [CrossRef]
  71. Kountouras, J.; Mylopoulos, N.; Chatzopoulos, D.; Zavos, C.; Boura, P.; Konstas, A.G.P.; Venizelos, J. Eradication of Helicobacter pylori May Be Beneficial in the Management of Chronic Open-Angle Glaucoma. Arch. Intern Med. 2002, 162, 1237. [Google Scholar] [CrossRef] [Green Version]
  72. Murphy, M.C.; Conner, I.P.; Teng, C.Y.; Lawrence, J.D.; Safiullah, Z.; Wang, B.; Bilonick, R.A.; Kim, S.G.; Wollstein, G.; Schuman, J.S.; et al. Retinal Structures and Visual Cortex Activity are Impaired Prior to Clinical Vision Loss in Glaucoma. Sci. Rep. 2016, 6, 31464. [Google Scholar] [CrossRef] [Green Version]
  73. Yu, L.; Xie, L.; Dai, C.; Xie, B.; Liang, M.; Zhao, L.; Yin, Y.; Wang, J. Progressive Thinning of Visual Cortex in Primary Open-Angle Glaucoma of Varying Severity. PLoS ONE 2015, 10, e0121960. [Google Scholar] [CrossRef] [Green Version]
  74. Han, M.L.; Chen, J.H.; Tsai, M.K.; Liou, J.M.; Chiou, J.M.; Chiu, M.J.; Chen, Y.C. Association between Helicobacter pylori infection and cognitive impairment in the elderly. J. Formos. Med. Assoc. 2018, 117, 994–1002. [Google Scholar] [CrossRef]
  75. Prevete, N.; Rossi, F.W.; Rivellese, F.; Lamacchia, D.; Pelosi, C.; Lobasso, A.; Necchi, V.; Solcia, E.; Fiocca, R.; Ceppa, P.; et al. Helicobacter pylori HP(2–20) Induces Eosinophil Activation and Accumulation in Superficial Gastric Mucosa and Stimulates VEGF-α and TGF-β Release by Interacting with Formyl-Peptide Receptors. Int. J. Immunopathol. Pharmacol. 2013, 26, 647–662. [Google Scholar] [CrossRef]
  76. Wang, X.L.; Zeng, J.; Feng, J.; Tian, Y.T.; Liu, Y.J.; Qiu, M.; Yan, X.; Yang, Y.; Xiong, Y.; Zhang, Z.H.; et al. Helicobacter pylori filtrate impairs spatial learning and memory in rats and increases Î2-amyloid by enhancing expression of presenilin-2. Front. Aging Neurosci. 2014, 6, 66. [Google Scholar] [CrossRef] [Green Version]
  77. Berrett, A.N.; Gale, S.D.; Erickson, L.D.; Brown, B.L.; Hedges, D.W. Helicobacter pylori moderates the association between 5-MTHF concentration and cognitive function in older adults. PLoS ONE 2018, 13, e0190475. [Google Scholar] [CrossRef] [Green Version]
  78. Bu, X.L.; Yao, X.Q.; Jiao, S.S.; Zeng, F.; Liu, Y.H.; Xiang, Y.; Xiang, C.R.; Liang, Q.H.; Wang, X.; Wang, H.Y.; et al. A study on the association between infectious burden and Alzheimer’s disease. Eur. J. Neurol. 2015, 22, 1519–1525. [Google Scholar] [CrossRef]
  79. Katsinelos, T.; Doulberis, M.; Polyzos, S.A.; Papaefthymiou, A.; Katsinelos, P.; Kountouras, J. Molecular Links Between Alzheimer’s Disease and Gastrointestinal Microbiota: Emphasis on Helicobacter pylori Infection Involvement. Curr. Mol. Med. 2019, 20, 3–12. [Google Scholar] [CrossRef]
  80. Boziki, M.; Grigoriadis, N.; Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A.; Kountouras, J. Potential impact of Helicobacter pylori-related Galectin-3 on chronic kidney, cardiovascular and brain disorders in decompensated cirrhosis. Dig. Liver Dis. 2020, 52, 121–123. [Google Scholar] [CrossRef]
  81. Boziki, M.; Polyzos, S.A.; Papaefthymiou, A.; Doulberis, M.; Bakirtzis, C.; Sintila, S.-A.; Touloumtzi, M.; Grigoriadis, N.; Kountouras, J. Potential impact of Helicobacter pylori-related metabolic syndrome and Galectin-3 on liver, chronic kidney and brain disorders. Metabolism 2021, 118, 154736. [Google Scholar] [CrossRef]
  82. Xie, J.; Cools, L.; van Imschoot, G.; van Wonterghem, E.; Pauwels, M.J.; Vlaeminck, I. Helicobacter pylori-derived outer membrane vesicles contribute to Alzheimer’s disease pathogenesis via C3-C3aR signalling. J. Extracell. Vesicles 2023, 12, 12306. [Google Scholar] [CrossRef] [PubMed]
  83. Franceschi, F.; Ojetti, V.; Candelli, M.; Covino, M.; Cardone, S.; Potenza, A.; Simeoni, B.; Gabrielli, M.; Sabia, L.; Gasbarrini, G.; et al. Microbes and Alzheimer’ disease: Lessons from H. pylori and GUT microbiota. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 426–430. [Google Scholar] [CrossRef] [PubMed]
  84. Llado-Saz, S.; Atienza, M.; Cantero, J.L. Increased levels of plasma amyloid-beta are related to cortical thinning and cognitive decline in cognitively normal elderly subjects. Neurobiol. Aging 2015, 36, 2791–2797. [Google Scholar] [CrossRef] [PubMed]
  85. Andrade-Guerrero, J.; Santiago-Balmaseda, A.; Jeronimo-Aguilar, P.; Vargas-Rodríguez, I.; Cadena-Suárez, A.R.; Sánchez-Garibay, C.; Molina, G.P.; Méndez-Catalá, C.F.; Delnas-Aguayo, M.d.C.; Diaz-Cintra, S.; et al. Alzheimer’s Disease: An Updated Overview of Its Genetics. Int. J. Mol. Sci. 2023, 24, 3754. [Google Scholar] [CrossRef]
  86. de Roeck, A.; van Broeckhoven, C.; Sleegers, K. The role of ABCA7 in Alzheimer’s disease: Evidence from genomics, transcriptomics and methylomics. Acta Neuropathol. 2019, 138, 201–220. [Google Scholar] [CrossRef] [Green Version]
  87. Narayan, P.; Sienski, G.; Bonner, J.M.; Lin, Y.-T.; Seo, J.; Baru, V.; Haque, A.; Milo, B.; Akay, L.A.; Graziosi, A.; et al. PICALM Rescues Endocytic Defects Caused by the Alzheimer’s Disease Risk Factor APOE4. Cell Rep 2020, 33, 108224. [Google Scholar] [CrossRef]
  88. Uddin, M.S.; Kabir, M.T.; Begum, M.M.; Islam, M.S.; Behl, T.; Ashraf, G.M. Correction to: Exploring the Role of CLU in the Pathogenesis of Alzheimer’s Disease. Neurotox. Res. 2020, 38, 1062. [Google Scholar] [CrossRef]
  89. Stage, E.; Duran, T.; Risacher, S.L.; Goukasian, N.; Do, T.M.; West, J.D.; Wilhalme, H.; Nho, K.; Phillips, M.; Elashoff, D.; et al. The effect of the top 20 Alzheimer disease risk genes on gray-matter density and FDG PET brain metabolism. Alzheimer’s Dement. Diagn. Assess. Dis. Monit. 2016, 5, 53–66. [Google Scholar] [CrossRef]
  90. Tesi, N.; van der Lee, S.J.; Hulsman, M.; Jansen, I.E.; Stringa, N.; van Schoor, N.; Meijers-Heijboer, H.; Huisman, M.; Scheltens, P.; Reinders, M.J.T.; et al. Centenarian controls increase variant effect sizes by an average twofold in an extreme case–extreme control analysis of Alzheimer’s disease. Eur. J. Hum. Genet. 2019, 27, 244–253. [Google Scholar] [CrossRef] [Green Version]
  91. You, M.; Zhou, X.; Yin, W.; Wan, K.; Zhang, W.; Li, C.; Li, C.; Li, M.; Zhu, W.; Zhu, X.; et al. The Influence of MTHFR Polymorphism on Gray Matter Volume in Patients with Amnestic Mild Cognitive Impairment. Front. Neurosci. 2021, 15, 778123. [Google Scholar] [CrossRef]
  92. Yuan, X.; Wang, H.; Zhang, F.; Zhang, M.; Wang, Q.; Wang, J. The common genes involved in the pathogenesis of Alzheimer’s disease and type 2 diabetes and their implication for drug repositioning. Neuropharmacology 2023, 223, 109327. [Google Scholar] [CrossRef]
  93. Cajavilca, C.E.; Gadhia, R.R.; Román, G.C. MTHFR Gene Mutations Correlate with White Matter Disease Burden and Predict Cerebrovascular Disease and Dementia. Brain Sci. 2019, 9, 211. [Google Scholar] [CrossRef] [Green Version]
  94. Kara, I.; Sazci, A.; Ergul, E.; Kaya, G.; Kilic, G. Association of the C677T and A1298C polymorphisms in the 5,10 methylenetetrahydrofolate reductase gene in patients with migraine risk. Mol. Brain Res. 2003, 111, 84–90. [Google Scholar] [CrossRef]
  95. Li, Y.; Laws, S.M.; Miles, L.A.; Wiley, J.S.; Huang, X.; Masters, C.L.; Gu, B.J. Genomics of Alzheimer’s disease implicates the innate and adaptive immune systems. Cell. Mol. Life Sci. 2021, 78, 7397–7426. [Google Scholar] [CrossRef]
  96. 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] [Green Version]
  97. Santarelli, L.; Gabrielli, M.; Cremonini, F.; Santoliquido, A.; Candelli, M.; Nista, E.C.; Pola, P.; Gasbarrini, G.; Gasbarrini, A. Atrophic gastritis as a cause of hyperhomocysteinaemia. Aliment. Pharmacol. Ther. 2004, 19, 107–111. [Google Scholar] [CrossRef]
  98. Hasan, T.; Arora, R.; Bansal, A.K.; Bhattacharya, R.; Sharma, G.S.; Singh, L.R. Disturbed homocysteine metabolism is associated with cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [Green Version]
  99. McCully, K.S. Homocysteine Metabolism, Atherosclerosis, and Diseases of Aging. Compr. Physiol. 2015, 6, 471–505. [Google Scholar] [CrossRef]
  100. Kountouras, J.; Polyzos, S.A.; Katsinelos, P.; Zeglinas, C.; Artemaki, F.; Tzivras, D.; Vardaka, E.; Gavalas, E.; Romiopoulos, I.; Symeonidou, C.; et al. Cardio-cerebrovascular disease and Helicobacter pylori-related metabolic syndrome: We consider eradication therapy as a potential cardio-cerebrovascular prevention strategy. Int. J. Cardiol. 2017, 229, 17–18. [Google Scholar] [CrossRef]
  101. Bostom, A.G.; Shemin, D.; Lapane, K.L.; Sutherland, P.; Nadeau, M.R.; Wilson, P.W.F.; Yoburn, D.; Bausserman, L.; Tofler, G.; Jacques, P.F.; et al. Hyperhomocysteinemia, hyperfibrinogenemia, and lipoprotein (a) excess in maintenance dialysis patients: A matched case-control study. Atherosclerosis 1996, 125, 91–101. [Google Scholar] [CrossRef]
  102. Román, G.C. Vascular dementia prevention: A risk factor analysis. Cerebrovasc. Dis. 2005, 20, 91–100. [Google Scholar] [CrossRef] [PubMed]
  103. Nieraad, H.; de Bruin, N.; Arne, O.; Hofmann, M.C.J.; Schmidt, M.; Saito, T.; Saido, T.C.; Gurke, R.; Schmidt, D.; Till, U.; et al. Impact of Hyperhomocysteinemia and Different Dietary Interventions on Cognitive Performance in a Knock-in Mouse Model for Alzheimer’s Disease. Nutrients 2020, 12, 3248. [Google Scholar] [CrossRef] [PubMed]
  104. Prats-Soteras, X.; Jurado, M.A.; Ottino-González, J.; García-García, I.; Segura, B.; Caldú, X.; Sánchez-Garre, C.; Miró, N.; Tor, C.; Sender-Palacios, M.; et al. Inflammatory agents partially explain associations between cortical thickness, surface area, and body mass in adolescents and young adulthood. Int. J. Obes. 2020, 44, 1487–1496. [Google Scholar] [CrossRef] [PubMed]
  105. Tan, B.; Venketasubramanian, N.; Vrooman, H.; Cheng, C.Y.; Wong, T.Y.; Ikram, M.K.; Chen, C.; Saima, H. Homocysteine and Cerebral Atrophy: The Epidemiology of Dementia in Singapore Study. J. Alzheimer’s Dis. 2018, 62, 877–885. [Google Scholar] [CrossRef] [PubMed]
  106. Cao, C.; Wang, Q.; Yu, H.; Yang, H.; Li, Y.; Guo, M.; Huo, H.; Fan, G. Morphological Changes in Cortical and Subcortical Structures in Multiple System Atrophy Patients with Mild Cognitive Impairment. Front. Hum. Neurosci. 2021, 15, 649051. [Google Scholar] [CrossRef]
  107. di Meco, A.; Li, J.G.; Praticò, D. Dissecting the Role of 5-Lipoxygenase in the Homocysteine-Induced Alzheimer’s Disease Pathology. J. Alzheimer’s Dis. 2018, 62, 1337–1344. [Google Scholar] [CrossRef] [Green Version]
  108. Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Chatzigeorgiou, S.; Katsinelos, P.; Grigoriadis, N.; Giartza-Taxidou, E.; Venizelos, I. Five-year survival after Helicobacter pylori eradication in Alzheimer disease patients. Cogn. Behav. Neurol. 2010, 23, 199–204. [Google Scholar] [CrossRef]
  109. Chen, Y.; Xu, C.; Xu, H.; Chen, W.; Wang, H.; Wang, Z.; Zhang, J. Persistent Helicobacter pylori infection for more than 3 years leads to elevated serum homocysteine concentration: A retrospective cohort study based on a healthy Chinese population. J. Gastroenterol. Hepatol. 2021, 36, 3077–3083. [Google Scholar] [CrossRef]
  110. Marino, M.C.A.; de Oliveira, C.A.; Rocha, A.M.C.; Rocha, G.A.; Clementino, N.C.D.; Antunes, L.F.; Oliveira, R.A.; Martins, A.S.; Del Puerto, H.L.; D’Almeida, V.; et al. Long-term effect of Helicobacter pylori eradication on plasma homocysteine in elderly patients with cobalamin deficiency. Gut 2007, 56, 469–474. [Google Scholar] [CrossRef] [Green Version]
  111. Franceschi, F.; Gasbarrini, A.; Polyzos, S.A.; Kountouras, J. Extragastric Diseases and Helicobacter pylori. Helicobacter 2015, 20, 40–46. [Google Scholar] [CrossRef] [Green Version]
  112. Chin, K.S.; Yassi, N.; Churilov, L.; Masters, C.L.; Watson, R. Prevalence and clinical associations of tau in Lewy body dementias: A systematic review and meta-analysis. Park. Relat. Disord. 2020, 80, 184–193. [Google Scholar] [CrossRef]
  113. Sohrabi, T.; Mirzaei-Behbahani, B.; Zadali, R.; Pirhaghi, M.; Morozova-Roche, L.A.; Meratan, A.A. Common Mechanisms Underlying α-Synuclein-Induced Mitochondrial Dysfunction in Parkinson’s Disease. J. Mol. Biol. 2023, 167992. [Google Scholar] [CrossRef]
  114. Rosca, E.; Simu, M. Parkinson’s Disease-Cognitive Rating Scale for Evaluating Cognitive Impairment in Parkinson’s Disease: A Systematic Review. Brain Sci. 2020, 10, 588. [Google Scholar] [CrossRef]
  115. Lee, H.M.; Koh, S.B. Many Faces of Parkinson’s Disease: Non-Motor Symptoms of Parkinson’s Disease. J. Mov. Disord. 2015, 8, 92–97. [Google Scholar] [CrossRef]
  116. Rong, S.; Li, Y.; Li, B.; Nie, K.; Zhang, P.; Cai, T.; Wang, L.; Zhang, Y. Meynert nucleus-related cortical thinning in Parkinson’s disease with mild cognitive impairment. Quant. Imaging Med. Surg. 2021, 11, 1554–1566. [Google Scholar] [CrossRef]
  117. Yoon, E.J.; Monchi, O. Probable REM sleep behavior disorder is associated with longitudinal cortical thinning in Parkinson’s disease. NPJ Park. Dis. 2021, 7, 19. [Google Scholar] [CrossRef]
  118. Wilson, H.; Niccolini, F.; Pellicano, C.; Politis, M. Cortical thinning across Parkinson’s disease stages and clinical correlates. J. Neurol. Sci. 2019, 398, 31–38. [Google Scholar] [CrossRef]
  119. Uribe, C.; Segura, B.; Baggio, H.C.; Abos, A.; Garcia-Diaz, A.I.; Campabadal, A.; Marti, M.J.; Valldeoriola, F.; Compta, Y.; Bargallo, N.; et al. Progression of Parkinson’s disease patients’ subtypes based on cortical thinning: 4-year follow-up. Park. Relat. Disord. 2019, 64, 286–292. [Google Scholar] [CrossRef]
  120. Fu, P.; Gao, M.; Yung, K.K.L. Association of Intestinal Disorders with Parkinson’s Disease and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. ACS Chem. Neurosci. 2020, 11, 395–405. [Google Scholar] [CrossRef]
  121. McGee, D.J.; Lu, X.-H.; Disbrow, E.A. Stomaching the Possibility of a Pathogenic Role for Helicobacter pylori in Parkinson’s Disease. J. Park. Dis. 2018, 8, 367–374. [Google Scholar] [CrossRef] [Green Version]
  122. Fan, X.; Zhang, L.; Li, H.; Chen, G.; Qi, G.; Ma, X.; Jin, Y. Role of homocysteine in the development and progression of Parkinson’s disease. Ann. Clin. Transl. Neurol. 2020, 7, 2332–2338. [Google Scholar] [CrossRef] [PubMed]
  123. Sampedro, F.; Martínez-Horta, S.; Horta-Barba, A.; Grothe, M.J.; Labrador-Espinosa, M.A.; Jesús, S.; Gómez, A.A.; Carrillo, F.; Puig-Davi, A.; Roldán Lora, F.; et al. Increased homocysteine levels correlate with cortical structural damage in Parkinson’s disease. J. Neurol. Sci. 2022, 434, 120148. [Google Scholar] [CrossRef] [PubMed]
  124. Periñán, M.T.; Macías-García, D.; Jesús, S.; Martín-Rodríguez, J.F.; Muñoz-Delgado, L.; Jimenez-Jaraba, M.V.; Buiza-Rueda, D.; Bonilla-Toribio, M.; Adarmes-Gómez, A.D.; Gómez-Garre, R.; et al. Homocysteine levels, genetic background, and cognitive impairment in Parkinson’s disease. J. Neurol. 2023, 270, 477–485. [Google Scholar] [CrossRef] [PubMed]
  125. Anamnart, C.; Kitjarak, R. Effects of vitamin B12, folate, and entacapone on homocysteine levels in levodopa-treated Parkinson’s disease patients: A randomized controlled study. J. Clin. Neurosci. 2021, 88, 226–231. [Google Scholar] [CrossRef]
  126. Czesnikiewicz-Guzik, M.; Bielanski, W.; Guzik, T.J.; Loster, B.; Konturek, S.J. Helicobacter pylori in the oral cavity and its implications for gastric infection, periodontal health, immunology and dyspepsia. J. Physiol. Pharmacol. 2005, 6, 77–89. [Google Scholar]
  127. Liu, Y.; Li, R.; Xue, X.; Xu, T.; Luo, Y.; Dong, Q. Periodontal disease and Helicobacter pylori infection in oral cavity: A meta-analysis of 2727 participants mainly based on Asian studies. Clin. Oral Investig. 2020, 24, 2175–2188. [Google Scholar] [CrossRef]
  128. Pisani, F.; Pisani, V.; Arcangeli, F.; Harding, A.; Singhrao, S.K. Locus Coeruleus Dysfunction and Trigeminal Mesencephalic Nucleus Degeneration: A Cue for Periodontal Infection Mediated Damage in Alzheimer’s Disease? Int. J. Environ. Res. Public Health 2023, 20, 1007. [Google Scholar] [CrossRef]
  129. Hu, S.; Li, S.; Ning, W.; Huang, X.; Liu, X.; Deng, Y.; Franceschi, D.; Ogbuehi, A.; Lethaus, B.; Savkovic, V.; et al. Identifying crosstalk genetic biomarkers linking a neurodegenerative disease, Parkinson’s disease, and periodontitis using integrated bioinformatics analyses. Front. Aging Neurosci. 2022, 14, 1032401. [Google Scholar] [CrossRef]
  130. Pareto, D.; Sastre-Garriga, J.; Auger, C.; Vives-Gilabert, Y.; Delgado, J.; Tintore, M.; Montalban, X.; Rovira, A. Juxtacortical lesions and cortical thinning in multiple sclerosis. Am. J. Neuroradiol. 2015, 36, 2270–2276. [Google Scholar] [CrossRef] [Green Version]
  131. Dubchenko, E.; Ivanov, A.; Spirina, N.; Smirnova, N.; Melnikov, M.; Boyko, A.; Gusev, E.; Kubatiev, A. Hyperhomocysteinemia and Endothelial Dysfunction in Multiple Sclerosis. Brain Sci. 2020, 10, 637. [Google Scholar] [CrossRef]
  132. Spirina, N.N.; Spirin, N.N.; Kiseleva, E.V.; Dubchenko, E.A.; Boyko, A.N. Homocysteine and markers of endothelial dysfunction in multiple sclerosis. Zhurnal Nevrol. I Psikhiatrii Im SS Korsakova 2021, 121, 90. [Google Scholar] [CrossRef]
  133. Oliveira, S.R.; Flauzino, T.; Sabino, B.S.; Kallaur, A.P.; Alfieri, D.F.; Kaimen-Maciel, D.R.; Morimoto, H.K.; Delicato de Almeida, E.R.; Lozovoy, M.A.B.; Vissoci Reiche, E.M.; et al. Elevated plasma homocysteine levels are associated with disability progression in patients with multiple sclerosis. Metab. Brain Dis. 2018, 33, 1393–1399. [Google Scholar] [CrossRef]
  134. Kaufmann, M.; Schaupp, A.L.; Sun, R.; Coscia, F.; Dendrou, C.A.; Cortes, A.; Kaur, G.; Evans, H.G.; Mollbrink, A.; Navarro, H.F.; et al. Identification of early neurodegenerative pathways in progressive multiple sclerosis. Nat. Neurosci. 2022, 25, 944–955. [Google Scholar] [CrossRef]
  135. Bayer, A.U.; Keller, O.N.; Ferrari, F.; Maag, K.-P. Association of glaucoma with neurodegenerative diseases with apoptotic cell death: Alzheimer’s disease and Parkinson’s disease. Am. J. Ophthalmol. 2002, 133, 135–137. [Google Scholar] [CrossRef]
  136. Kountouras, J.; Mylopoulos, N.; Boura, P.; Bessas, C.; Chatzopoulos, D.; Venizelos, J.; Zavos, C. Relationship between Helicobacter pylori infection and glaucoma. Ophthalmology 2001, 108, 599–604. [Google Scholar] [CrossRef]
  137. Kountouras, J.; Zavos, C.; Chatzopoulos, D. Primary open-angle glaucoma: Pathophysiology and treatment. Lancet 2004, 364, 1311–1312. [Google Scholar] [CrossRef]
  138. Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A.; Bargiotas, P.; Liatsos, C.; Shiva Srivastava, D.; Zavos, C.; Katsinelos, P.; Kountouras, J. Association between active Helicobacter pylori infection and glaucoma: A systematic review and meta-analysis. Microorganisms 2020, 8, 894. [Google Scholar] [CrossRef]
  139. Yu, L.; Xie, B.; Yin, X.; Liang, M.; Evans, A.C.; Wang, J.; Dao, C. Reduced Cortical Thickness in Primary Open-Angle Glaucoma and Its Relationship to the Retinal Nerve Fiber Layer Thickness. PLoS ONE 2013, 8, e73208. [Google Scholar] [CrossRef] [Green Version]
  140. Xiao, H.; Wang, J.; Barwick, S.R.; Yoon, Y.; Smith, S.B. Effect of long-term chronic hyperhomocysteinemia on retinal structure and function in the cystathionine-β-synthase mutant mouse. Exp. Eye Res. 2022, 214, 108894. [Google Scholar] [CrossRef]
  141. Navneet, S.; Zhao, J.; Wang, J.; Mysona, B.; Barwick, S.; Ammal Kaidery, N.; Kaddour-Djebbar, S.A.I.; Bollag, W.B.; Thomas, B. Hyperhomocysteinemia-induced death of retinal ganglion cells: The role of Müller glial cells and NRF2. Redox. Biol. 2019, 24, 101199. [Google Scholar] [CrossRef]
  142. Papaefthymiou, A.; Doulberis, M.; Katsinelos, P.; Liatsos, C.; Polyzos, S.A.; Kotronis, G.; Papanikolaou, K.; Kountouras, J. Impact of nitric oxide’s bidirectional role on glaucoma: Focus on Helicobacter pylori-related nitrosative stress. Ann. N. Y. Acad. Sci. 2020, 1465, 10–28. [Google Scholar] [CrossRef] [PubMed]
  143. Jünemann, A.; Rejdak, R.; Hohberger, B. Stellenwert vom Homocystein beim Glaukom. Klin. Monbl. Augenheilkd. 2018, 235, 163–174. [Google Scholar] [CrossRef] [PubMed]
  144. Türkcü, F.M.; Köz, Ö.G.; Yarangümeli, A.; Öner, V.; Kural, G. Plasma homocysteine, folic acid, and vitamin B12 levels in patients with pseudoexfoliation syndrome, pseudoexfoliation glaucoma, and normotensive glaucoma. Medicina 2013, 49, 214–218. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, Z.; Zhu, W.; Xing, Y.; Jia, J.; Tang, Y. B vitamins and prevention of cognitive decline and incident dementia: A systematic review and meta-analysis. Nutr. Rev. 2022, 80, 931–949. [Google Scholar] [CrossRef] [PubMed]
  146. Besteher, B.; Wagner, G.; Koch, K.; Schachtzabel, C.; Reichenbach, J.R.; Schlösser, R.; Sauer, H.; Schultz, C.C. Pronounced prefronto-temporal cortical thinning in schizophrenia: Neuroanatomical correlate of suicidal behavior? Schizophr. Res. 2016, 176, 151–157. [Google Scholar] [CrossRef]
  147. Zhilyaeva, T.v.; Piatoikina, A.S.; Bavrina, A.P.; Kostinal, O.v.; Zhukova, E.S.; Shcherbatyuk, T.G.; Blagonravova, A.S.; Dubinina, E.E.; Mazo, G.E. Homocysteine in Schizophrenia: Independent Pathogenetic Factor with Prooxidant Activity or Integral Marker of Other Biochemical Disturbances? Schizophr. Res. Treatment 2021, 2021, 7721760. [Google Scholar] [CrossRef]
  148. Rasool, M.; Malik, A.; Saleem, S.; Ashraf, M.A.B.; Khan, A.Q.; Waquar, S.; Zahid, A.; Shaheen, S.; Abu-Elmagd, M.; Gauthaman, K.; et al. Role of Oxidative Stress and the Identification of Biomarkers Associated With Thyroid Dysfunction in Schizophrenics. Front. Pharmacol. 2021, 12, 6946287. [Google Scholar] [CrossRef]
  149. Kleine, A.D.; Reuss, B. Interactions of Antibodies to the Gram-Negative Gastric Bacterium Helicobacter pylori with the Synaptic Calcium Sensor Synaptotagmin 5, Correlate to Impaired Vesicle Recycling in SiMa Human Neuroblastoma Cells. J. Mol. Neurosci. 2021, 71, 481–505. [Google Scholar] [CrossRef]
  150. Yilmaz, Y.; Gul, C.B.; Arabul, M.; Eren, M.A. Helicobacter pylori: A role in schizophrenia? Med. Sci. Monit. 2008, 14, HY13-6. [Google Scholar]
  151. Franceschi, F.; Annalisa, T.; Teresa, D.R.; Giovanna, D.; Ianiro, G.; Franco, S.; Gerardi, V.; Tesori, V.; Lopetuso, L.R.; Gasbarrini, A. Role of Helicobacter pylori infection on nutrition and metabolism. World J. Gastroenterol. 2014, 20, 12809–12817. [Google Scholar] [CrossRef]
  152. Moser, P. Evaluating negative-symptom-like behavioural changes in developmental models of schizophrenia. Eur. Neuropsychopharmacol. 2014, 24, 774–787. [Google Scholar] [CrossRef]
  153. Brown, H.E.; Roffman, J.L. Vitamin supplementation in the treatment of schizophrenia. CNS Drugs 2014, 28, 611–622. [Google Scholar] [CrossRef]
  154. Cai, X.; Li, X.; Jin, Y.; Zhang, M.; Xu, Y.; Liang, C.; Weng, Y.; Yu, W.; Li, X. Vitamins and Helicobacter pylori: An Updated Comprehensive Meta-Analysis and Systematic Review. Front. Nutr. 2022, 8, 781333. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of proposed pathophysiological pathways connecting Helicobacter pylori infection and neurodegeneration. AD, Alzheimer’s disease; α-syn, α-synuclein; BBB, blood–brain barrier; Gal-3, Galectin-3; H. pylori, Helicobacter pylori; HHcy, hyperhomocysteinemia; OMV, outer membrane vehicles; TNFα, tumor necrosis factor-α.
Figure 1. Schematic illustration of proposed pathophysiological pathways connecting Helicobacter pylori infection and neurodegeneration. AD, Alzheimer’s disease; α-syn, α-synuclein; BBB, blood–brain barrier; Gal-3, Galectin-3; H. pylori, Helicobacter pylori; HHcy, hyperhomocysteinemia; OMV, outer membrane vehicles; TNFα, tumor necrosis factor-α.
Medicina 59 00504 g001
Figure 2. Schematic illustration of proposed pathophysiological mechanism (s) connecting Helicobacter pylori infection and neurodegeneration through the hyperhomocysteinemia pathway. CCV, cardio-cerebrovascular disorders; H. pylori, Helicobacter pylori.
Figure 2. Schematic illustration of proposed pathophysiological mechanism (s) connecting Helicobacter pylori infection and neurodegeneration through the hyperhomocysteinemia pathway. CCV, cardio-cerebrovascular disorders; H. pylori, Helicobacter pylori.
Medicina 59 00504 g002
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

Kountouras, J.; Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A.; Zavos, C.; Kazakos, E.; Arapoglou, S.; Kyrailidi, F.; Mouratidou, M.C.; Boziki, M.; et al. Controlling the Impact of Helicobacter pylori-Related Hyperhomocysteinemia on Neurodegeneration. Medicina 2023, 59, 504. https://doi.org/10.3390/medicina59030504

AMA Style

Kountouras J, Doulberis M, Papaefthymiou A, Polyzos SA, Zavos C, Kazakos E, Arapoglou S, Kyrailidi F, Mouratidou MC, Boziki M, et al. Controlling the Impact of Helicobacter pylori-Related Hyperhomocysteinemia on Neurodegeneration. Medicina. 2023; 59(3):504. https://doi.org/10.3390/medicina59030504

Chicago/Turabian Style

Kountouras, Jannis, Michael Doulberis, Apostolis Papaefthymiou, Stergios A. Polyzos, Christos Zavos, Evangelos Kazakos, Stergios Arapoglou, Foteini Kyrailidi, Maria C. Mouratidou, Marina Boziki, and et al. 2023. "Controlling the Impact of Helicobacter pylori-Related Hyperhomocysteinemia on Neurodegeneration" Medicina 59, no. 3: 504. https://doi.org/10.3390/medicina59030504

Article Metrics

Back to TopTop