Next Article in Journal
Suppressive Effects of 4-(Phenylsulfanyl) Butan-2-One on CCL-1 Production via Histone Acetylation in Monocytes
Next Article in Special Issue
Piperazine Derivative Stabilizes Actin Filaments in Primary Fibroblasts and Binds G-Actin In Silico
Previous Article in Journal
Anti-Inflammatory Activity of Bilberry (Vaccinium myrtillus L.)
Previous Article in Special Issue
Analysis of SOD2 rs4880 Genetic Variant in Patients with Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Implications of Microorganisms in Alzheimer’s Disease

1
Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
2
Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Korea
3
Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India
4
Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, Punjab, India
5
Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun 248007, Uttarakhand, India
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Issues Mol. Biol. 2022, 44(10), 4584-4615; https://doi.org/10.3390/cimb44100314
Submission received: 6 September 2022 / Revised: 23 September 2022 / Accepted: 28 September 2022 / Published: 30 September 2022

Abstract

:
Alzheimer’s disease (AD) is a deadly brain degenerative disorder that leads to brain shrinkage and dementia. AD is manifested with hyperphosphorylated tau protein levels and amyloid beta (Aβ) peptide buildup in the hippocampus and cortex regions of the brain. The nervous tissue of AD patients also contains fungal proteins and DNA which are linked to bacterial infections, suggesting that polymicrobial infections also occur in the brains of those with AD. Both immunohistochemistry and next-generation sequencing (NGS) techniques were employed to assess fungal and bacterial infections in the brain tissue of AD patients and non-AD controls, with the most prevalent fungus genera detected in AD patients being Alternaria, Botrytis, Candida, and Malassezia. Interestingly, Fusarium was the most common genus detected in the control group. Both AD patients and controls were also detectable for Proteobacteria, followed by Firmicutes, Actinobacteria, and Bacteroides for bacterial infection. At the family level, Burkholderiaceae and Staphylococcaceae exhibited higher levels in the brains of those with AD than the brains of the control group. Accordingly, there is thought to be a viscous cycle of uncontrolled neuroinflammation and neurodegeneration in the brain, caused by agents such as the herpes simplex virus type 1 (HSV1), Chlamydophila pneumonia, and Spirochetes, and the presence of apolipoprotein E4 (APOE4), which is associated with an increased proinflammatory response in the immune system. Systemic proinflammatory cytokines are produced by microorganisms such as Cytomegalovirus, Helicobacter pylori, and those related to periodontal infections. These can then cross the blood–brain barrier (BBB) and lead to the onset of dementia. Here, we reviewed the relationship between the etiology of AD and microorganisms (such as bacterial pathogens, Herpesviridae viruses, and periodontal pathogens) according to the evidence available to understand the pathogenesis of AD. These findings might guide a targeted anti-inflammatory therapeutic approach to AD.

1. Introduction

Alzheimer’s disease (AD) is a deadly neurodegenerative illness that mostly impacts the elderly and is a major health concern for the geriatric population worldwide. AD prevalence increases substantially with age, reaching 50% in 85-year-olds [1]. AD is expected to become significantly more common in the geriatric population as the median life expectancy grows. The worldwide incidence of dementia is about 24 million, and it is estimated to surge fourfold by the year 2050 [2]. As such, there is a need for modern treatment options given the probable risk factors for AD and treatments that can delay AD onset and occurrence [3].
About 5000 species of gut microorganisms, notably Firmicutes, Bacteriodetes, Actinobacteria, and Proteobacteria, have been reported in the human intestinal lumen. These microbes play crucial roles in gut digestion and absorption functions [4]. Among these microorganisms, in particular, Chlamydia pneumoniae and Spirochetes have multiple research findings pointing to their significance in the pathogenesis of AD [5].
According to the AD pathogen hypothesis, pathogens operate as triggers, in conjunction with genetic variables, in starting the accumulation and/or processing of Aβ, hyperphosphorylated tau proteins, and inflammation in the brains of AD patients [6]. HSV1 and other pathogens, such as C. pneumoniae and Spirochetes, are commonly found in the brains of AD patients, as these may evade the host immune response and infect the brain [7,8]. In vitro and animal studies have demonstrated that pathogens facilitate amyloid plaque formation and increased levels of hyperphosphorylated tau [9,10]. Such pathogens induce a glial inflammatory response, damaging and killing neurons directly and indirectly [11]. In the brains of AD patients, there are major inflammatory cascades [12,13], and these pathways combine to cause further neurodegeneration and disease progression.
This review examines the evidence relating to the Herpesviridae, HSV1, and CMV, and the bacterial pathogens C. pneumoniae, Spirochetes, periodontal pathogens, and Helicobacter pylori, and their relation to AD etiology. Regarding EBV and HHV6, the Herpesviridae EBV has minimal evidence of possibly contributing to AD etiology. The possible role of poisons or other environmental cofactors in the pathogenesis of AD was studied elsewhere [14]. These factors cannot be ruled out as a contributor to AD onset and progression.

2. AD Etiology

Identifying the contributors to AD in the human brain is a contentious topic (Figure 1); however, microorganisms appear to play a role in the progression of AD. According to evidence obtained from the postmortem examination of more than 1000 affected individuals [15], there could be a variety of factors that cause AD, and, according to a number of scientists, the primary variables in AD are the levels of Aβ and the tau protein. Aβ is produced by the APP gene, located on chromosome 2 [16], and large amounts of Aβ plaques are present not only in AD patients but also in healthy individuals. However, aggregation of Aβ and neurofibrillary tangles formed by hyperphosphorylated tau protein in the cortical and limbic areas of the human brain contribute to the pathogenesis of AD [17]. As hallmarks of AD, plaque and tangles are formed due to enzymatic excision of APP by enzymes and secretases [18,19]; these are linked to amyloidosis, inflammation, and significant synaptic brain abnormalities [20,21]. The gradual accumulation of the tau protein or Aβ aggregates or their mislocalization in unhealthy neurons leads to disproportionate proteostasis. These events impact synaptic terminal configuration and synaptic activity, eventually leading to synapse loss [22,23]. Furthermore, synaptic dysfunction is directly linked to a decline in cognitive function of the brain [24,25].

2.1. Role of Bacteria

Seropositive response to Borreliabur gdorferi (B. burgdorferi) was detected in both AD patients and healthy controls, but the detection rate was higher in AD patients and the onset risk of AD in seropositive IgG patients to B. burgdorferi. Therefore, B. burgdorferi may act as a risk factor for AD [26]. A previous study reported that the expression levels of Aβ and phospho-tau proteins increase after infection of B. burgdorferi in vitro, and B. burgdorferi can be detected in the brain tissues of AD patients [27]. Glial and neuronal cells have also been shown to generate amyloid precursor protein (APP) and p-tau when introduced to B. burgdorferi [9]. B. burgdorferi was originally discovered in blood and the CSF of roughly 10 infected AD and Parkinson’s patients following their brain autopsy [27,28,29]. B. burgdorferi was also found in studies to process Aβ precursor protein in glial and neuronal cells [3]. In the brain tissue of individuals with and without dementia, T. denticola, Tannerella forsythia, and P. gingivalis were observed [30]. Interestingly, the levels of LPS from P. gingivalis were considerably greater in the brain tissue of AD patients compared to non-AD control tissues [31]. There was also an association of the presence of Spirochetes and AD, with Spirochetes being found in over 90% of AD patient brain samples, while B. burgdorferi was found in roughly 25.3% using PCR and antibody detection methods. Systemically, Spirochetes and Treponema denticola are the most common isolated bacteria in moderate-to-severe periodontitis [32], and individuals with AD have been reported to contain periodontopathic bacteria, indicating that these microorganisms can enter the brain, possibly through peripheral nerves and the circulatory system [33]. Spirochete bacteria are also often associated with syphilitic dementia, which results in the deposition of gray matter inside the cerebral and other regions of the brain. In such instances, numerous studies have discovered an increase in spirochete colonies in the cerebral brain, primarily the treponema palladium [34]. The prevalence of oral treponemas in the trigeminal ganglia supports the notion of microbial invasion via neural pathways [35,36], and the presence of large bacterial plaques are generally considered indicators of oral bacteria in the systemic circulation. Microbes have also been linked to various neuroinflammatory and immunological conditions, and studies have shown that neuroinflammation plays a key role in AD. The brain–gut axis, also known as the brain–gut–microbiome axis, is a reversible relationship among the brain, gut, and gut bacteria [37].

2.2. Role of Viruses

Several studies have also linked HSV1 and Herpesviridae members, such as Epstein–Barr virus (EBV), Cytomegalovirus (CMV), and HHV6, to AD occurrence [38,39,40,41,42]. For HSV1/2 and EBV, such viruses are capable of latent residence in the peripheral nervous system, and they target the same parts of the central nervous system, namely, the temporoparietal cortex and hippocampus, impacted in AD and in acute cases of encephalitis (for HSV1/2 and EBV) [43]. The Herpes simplex viruses such as HSV6, HSV7, HSV8, Varicella-Zoster virus (VZV) (HSV3), EBV (HSV4), and CMV (HSV5) are neurotrophic and neuroinvasive with double-stranded DNA belonging to the Herpesviridae family [44]. Among this group, HSV1 has been demonstrated to target the trigeminal ganglia or olfactory neurons in the peripheral nervous system [45,46], and HSV1 has been detected in the brain samples, particularly for the APOE4 genotype with susceptibility to AD [43]. HSV1 incidence also includes 3.7 billion people worldwide aged 50 or older, or 67% of the population [47]. The levels of amyloidosis in the AD brain samples also correlate with levels of HSV1, implying that HSV1 is involved in the progression of AD [48]. Herpes simplex virus type 1 (HSV1) is the infection most commonly related to amyloidosis in AD patients [49,50,51,52]. Proteins from the Herpesviridae family have been found to interact with products of multiple AD susceptibility genes, namely, apolipoprotein E (APOE), phosphatidylinositol-binding clathrin assembly protein (PICALM), complement receptor 1 (CR1), and clusterin (CLU), which are associated with dementia [7,53,54]. It was found that HSV1 upregulates amyloid beta (Aβ) generation, and Herpesviridae DNA signals colocalize with amyloid plaques in AD samples [7]. The activity of Aβ with respect to the presence of neurotropic Herpesviridae virus HSV1, human herpes virus 6A (HHV6A), and human herpes virus 6B (HHV6B) was demonstrated. Herpesviridae are common human infections, with neuro-infection rates surpassing 100% in the general population [55]. Between extended periods of dormancy, HSV1 undergoes acute and episodic reactivation. HHV6A and HHV6B are also implicated in an expanding number of chronic inflammatory illnesses with foci inside and outside the CNS due to their ongoing low levels of replication [44]. Researchers have also discovered an increased abundance of Roseoloviruses HHV6 and HHV7, as well as HSV1, in the brains of AD patients [56]. Following the effects of such infections, extensive beta production with Aβ fibrils and neuroinflammation has been shown in 2D and 3D brain models of the human brain using human-induced pluripotent stem cells (hiPSCs) and HSV1 infection [45].

2.3. Role of Fungi

Fungal presence has also been detected via a Fungitell test for the blood samples obtained from AD patients who had Aβ deposition, leading to neurofibrillary tangles (NFTs), according to Alonso and colleagues [57]. It has also been observed that consuming antifungal medications reduces the incidence of dementia [58]. It is known that fungal infections can result in inflammatory responses, in addition to bringing vascular changes [59]. Brain samples from 11 AD patients out of a total of 29 showed strong positivity to most Candida species examined, while two more patients showed high detection of a single Candida species. Candida famata and Candida albicans are also both gut microbes, with Candida albicans having also been reported on vaginal surfaces [60].

2.4. AD and Periodontis

Periodontitis is a disorder caused by bacteria such as Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, Treponema denticola, and Tannerella forsythia, which cause inflammation affecting tooth support and promoting gum damage. There is also gingivitis caused by P. gingivalis bacteria, which can lead to chronic systemic inflammation, as well as bone and soft-tissue degeneration in the jaw, also leading to tooth loss [33,61,62,63]. AD and periodontitis carry comparable risk factors. In plaque-induced gingivitis, half of the bacteria are Gram-positive; however, as the extent of inflammation increases, the number of Gram-negative bacteria also increases. Given subgingival dysbiosis with Porphyromonas gingivalis, the keystone pathogen of periodontal disease, shown to relate to AD occurrence, a hypothesis has been put forward in explaining the cause of AD along the lines of its two hallmark proteins for AD (Aβ fibrils and phosphorylated-tau tangles). It has been noted that advancement in age and loss of up to nine teeth are the primary risk factors for sporadic AD linked to chronic periodontitis [39]. Approximately 85% of bacteria in periodontitis are Gram-negative with Aggregatibacter actinomycetemcomitans (Actinobacillus actinomycetemcomitans), Tannerella forsythensis, P. gingivalis, and T. denticola considered the prime periodontal pathogens [64,65]. The Gram-negative bacterium, Helicobacter pylori, which is repressed by inhibition of GSK-3, causes inflammation and neurotoxicity by upregulating tau phosphorylation [66]. A link between periodontal disease and amyloid buildup in the brains of healthy elderly subjects was discovered using positron emission tomography (PET) imaging [65].

2.5. Other Factors Responsible for AD

Another prominent cause of AD is the plaque formation outside of the neuron as part of the AD cascade hypothesis with the aberrant formation of APP cleavage products due to the beta- and gamma-secretase cleavage of APP C-terminal residues, leading to the formation of APP83 and APP99 cleavage products. In a healthy individual, however, APP C-terminal cleavage occurs with the help of alpha and gamma-secretase, thought to facilitate immune stimulation and inflammation in the central nervous system [17].
Although it is still unclear if microorganisms such as bacteria, fungi, and viruses play a role in neurological diseases, we can claim that microbes are associated with neuro-inflammatory and immunological responses on the basis of multiple research findings. Host bacteria are primarily found as a part of human gut microbiota for digestive tract symbiosis. Periodontal bacteria, Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria, are all examples of such gut bacteria. In addition, viruses from the Herpesviridae and Retroviridae families, particularly the HSV, have been linked to AD. As previously noted, fungi such as Candida spp., Cladosporium, Rhodotorula mucilaginosa, Malassezia sp., Saccharomyces cerevisiae, and Penicillium may also be implicated in neurological disease [67]. Experimental findings include viruses and bacterial genetic material found in multiple AD patients’ brains.
Inflammation in the brain is thought to be brought about by two independent mechanisms: first, a loss of BBB in the systemic circulation, and second, agents that affect certain neuronal pathways [68,69]. An example of such agents is lipopolysaccharide (LPS) [70]. Upon binding of LPS to LPS-binding protein (LBP), LPS can signal to CD14 receptor-positive cells [71]. In these cells, LPS binds to the CD14/TLR4/MD4 receptor complex, causing the secretion of cytokines such as TNF, IL-1, IL-6, and IL-8, which in turn promote the production of prostaglandins and leukotrienes that cause inflammation and the onset of septic shock promoted by monocyte and macrophages. There is also the release of histamine when LPS binds to the CD14/TLR4/MD4 receptor complex, leading to vasodilation and stimulation of the coagulation cascade [72].
In 1998, Balin and colleagues examined patients with diverse medical histories, some of whom died from cancer, cardiac arrest, multiorgan failure, respiratory failure, systemic infection, and other severe diseases, with late-onset AD confirmed in their autopsy through histopathologic examination by a certified neuropathologist. PCR, electron, and immunoelectron microscopy of tissues for relevant AD brain structures also revealed chlamydial infection with transcriptional activity in neuropathology regions of the brains of AD patients, indicating that bacterial infection may be a significant predictor of late-onset AD. Samples were analyzed from diverse brain regions that display AD-related neuronal loss (e.g., hippocampus and temporal cortex), and a zone often mildly affected or unscathed by AD, i.e., the cerebellum. In addition, C. pneumoniae (Chlamydiaceae family) transcripts were detected by RT-PCR; this family of organisms also causes infections in the respiratory system and lungs [73]. Pneumonia is a general cause of death in AD patients [74]. Yeast is a strong model for basic cellular and molecular biology research, and 31% of 6000 genes are preserved between yeast and human species [75,76]. In addition, S. cerevisiae is mostly used as a major yeast model for AD study because it is suitable for expression studies of Aβ and tau protein [77,78].

3. Gut Microbiota and AD

Several gut bacteria are responsible for controlling and sustaining the host’s health [79]. Such a microbial community helps regulate neurotransmitter metabolism, form biological substances in the gut under normal physiological conditions, and maintain a healthy and balanced environment known as eubiosis [37,80]. Disruption of this ecosystem, such as via excessive antibiotic use, immune system suppression, and changes in gastrointestinal barriers, can lead to disorders including dysbiosis, which is associated with conditions such as inflammatory bowel disease (IBD), obesity, allergies, type 1 diabetes mellitus, autism, and colorectal cancer [79,81]. Microbes in the gut, such as bacteria and viruses, have been documented as pathogens responsible for various acute and chronic disorders, impacting various organs by various means from their site of infection [7,79,82].
The possibility of potential therapeutic interventions increases as a pivotal role for the microbiome in the etiology of AD, and a close interrelation with the gut is found. Dysbiosis is characterized by an imbalance in the microbial population, leading to a shortage of microbiome diversity in the digestive tract or an enhanced intestinal permeability [83]. The discovered interactions between the intestine’s enteric nervous system and the central nervous system have established a link between gut bacteria and brain physiology and function [37,79]. Various biological molecules are exchanged between these two systems as they flow through the bloodstream and pass through the gut mucosa and the BBB. Neurotoxic agents such as D-lactic acid and ammonia produced by the gut bacteria can potentially cause neuronal injury [84], and the release of proinflammatory substances such as cytokines and immune activators capable of generating neuro-inflammation can start an inflammatory process in the brain [85,86]. With an imbalance in gut microbiota metabolic activities, there is also a causal component for anxiety, depression, cognitive impairment, learning, and behavioral issues seen in various neurodegenerative disorders, including AD (Figure 2) [79,84,87].

4. Effects of Bacterial and Viral Infection on AD

According to recent studies, gut bacteria may have a role in AD development (Table 1) [88,89,90]. Various research methodology has been utilized to identify the microorganisms associated with AD progression (Figure 3). Lactobacillus species, Bifidobacteria (Actinobacteria), Verrucomicrobia, Spirochetes, Proteobacteria, Fusobacteria, Firmicutes, and Cyanobacteria are among the bacteria that make up the gut microbial community [88], and there are signs that bacterial infection may initiate some of the degenerative processes linked to AD. Bacterial infections such as Helicobacter pylori [91], B. burgdorferi, Chlamydia pneumonia [92], Escherichia coli, Shigella, Eubacterium rectale [93], and Bacteriodesfragilis have all been linked to AD, with some, however, working together to reduce their infection load in AD patients. The production of neurotoxic chemicals is one route via which these bacteria might cause their harmful effects. For example, in the central nervous system, Lactobacillus species and actinobacteria may metabolize glutamate to create gamma-aminobutyric acid (GABA) [88,94]. Such elevated levels of GABA levels in the gut can impact the GABA levels in the central nervous system, and high amounts of GABA in neuronal cells are linked to memory loss, depression, and synaptic disruption [88,95].
Inflammatory activators, Aβ peptides, and other neurotoxic chemicals are also released by gut bacteria, which can impair the host immune system [57,92]. These bacteria can create endotoxins, such as lipopolysaccharides, typically present in the outer membrane of Gram-negative bacteria [96]. Gram-negative bacteria that produce LPS are abundant in the stomach, saliva, respiratory and urinary tract, skin, dental plaques, and lungs [97]. Concentrations of LPS rise following a bacterial infection or changes in the metabolic activities of gut microbiota caused by inflammation, which in turn can lead to neurodegeneration [86,97]. Regarding the functional effects of LPS on the CNS, previous research has shown that LPS causes microglial activation, memory difficulties, and overall neural damage in mice [98]. LPS also stimulates Aβ aggregation and amyloid plaque formation, which are key pathogenic processes in the progression of AD [96]. Miklossy and colleagues discovered the existence of amyloid plaques that resembled the plaques in mice following the inhalation of C. pneumoniae. Spirochetes were also found to cause amyloid-like symptoms in neuronal and glial cells, which might be connected to neurodegeneration [99].
Viruses, particularly the HSV and the cytomegalovirus have been linked to AD [10,100]. Inflammatory signals produced by viral infections may harm neurons, resulting in neurodegeneration [43]. For example, HSV1 has been demonstrated to cause the generation of Aβ peptide and an increase in tau phosphorylation, which can lead to Aβ peptide aggregation, the development of amyloid plaques, and neurofibrillary tangles in cell and animal models [10,101]. HSV1 was documented to cause the development of Aβ peptides (A40 and A42) in human neuroblastoma cells. HSV1 also caused hyperexcitability and increased intracellular calcium signals in rat cortical neurons, resulting in modification of the amyloid precursor processing pathway and an aberrant rise in Aβ formation [10,42]. Another example is HHV6 found in the hippocampus, frontal and temporal cortex, and hippocampus of AD patients [49]. Despite the evidence that the HHV6 virus is not directly linked to AD, investigations have revealed that it augments the harm produced by the HSV1 virus and may create brain lesions in dually infected individuals [101].

5. Animal Models Used to Identify a Role for Various Microorganisms in AD

5.1. Mouse Models

Various mouse models have been used to define the role of microorganisms in AD progression, and they are listed in Table 2. Examples of the experiments shown assayed activation changes in the brain of APOE−/− mice, alterations in microbiome composition, changes in Aβ deposition, and changes in gliosis surrounding Aβ plaques. The experimental mice were either treated with antibiotics or Gram-negative oral anaerobes (Porphyromonas gingivalis). In another study by Brandscheid et al., changes in the fecal microbiota with age in 5XFAD mice were correlated with changes in trypsin reduction in fecal proteins and changes in human APP expressed not only in the brain but also in the gut tissue [118]. Another study with probiotic treatment in 3XTg-AD mice correlated changes in the plasma concentration of inflammatory cytokines and gut hormones with an accumulation of Aβ aggregates and brain damage [119].

5.2. Drosophila Models

Drosophila has been used in various studies to identify a genetic disposition for genes related to the disease of interest. Drosophila has emerged as a model system for understanding neurodegeneration pathways in various severe neurodegenerative models. These disease models are also suitable for in vivo testing of various therapeutic drug candidates. The genetic research employing these animal models has revealed new insights into neurodegenerative processes. We believe that further investigation of these animal models will further our understanding of neurodegeneration and allow for the development of novel therapeutics for severe degenerative disorders [130]. As an example, Wu et al. used transgenic Drosophila to provide evidence of the involvement of microorganisms in AD progression. In their experiments, researchers subjected Drosophila flies to Enterobacteria infection, resulting in progressive AD by increased immunological hemocyte recruitment to the brain. Interestingly, genetic hemocyte depletion also attenuated neuroinflammation and reduced neurodegeneration [128].

6. Potential Therapeutic Approaches in AD

6.1. Fecal Microbiota Transplantation

The delivery of fecal material from a healthy donor to the gastrointestinal tract of a diseased individual is known as fecal microbiota transplantation (FMT). This is an effective method of altering gut flora. Following the repeated outbreaks of severe Clostridioides difficile infection (CDI) in North America and Europe, caused partly by exposure to the Clostridioides hypervirulent NAP1/B1/027 strain, FMT has become popular in the recent decade for addressing gut flora anomalies. Cure rates of more than 90% in difficult-to-treat patients were recorded in randomized, controlled trials demonstrating the efficacy of FMT for severe CDI [131]. Improvements in a variety of extraintestinal diseases, including autism spectrum disorder (ASD), multiple sclerosis (MS), and myoclonus dystonia, have also been documented for FMT [132].
Several recent investigations have found a substantial correlation between gut microbiome changes and cognitive behavior [133,134]. Using 16S rRNA gene sequencing, researchers analyzed and compared variations in the makeup of the gut microbiota in AD mice models (APP, presenilin 1 (PS1), and age-matched healthy wildtype (WT) mice) [95]. Compared to the WT group, APP and PS1 mice contained considerably fewer bacteria, proteobacteria, and actinomycetes but significantly more Bacteroidetes and Trichomonas [135]. Furthermore, germ-free mice had considerably higher brain levels of Aβ after receiving fecal samples from AD animals; however, fecal transplants from WT mice had no influence on the behavioral or health outcomes of the affected mice [136].

6.2. Beta-Secretase

The major therapeutic molecular targets are beta-secretase (BACE1) [137,138] and gamma-secretase [139], involved in the processing of APP, and tau proteins [140], involved in the breakdown of the microtubules that keep the axonal routes together [141,142]. Beta-secretase is the most researched protein in AD (Table 2), and its protease activity causes plaque development [142]. It splits APP into A40 and A42 fragments [143], with the latter playing a key role in Familial AD. BACE1 has a cytoplasmic tail that aids in molecular maturation and intracellular trafficking, and its inhibition can slow down plaque deposition between neurons [143]. This enzyme contains an aspartate residue as an active-site moiety [144], which is also implicated in dimer and myelin sheath production outside the cell [143]. The endoplasmic reticulum forms the immature BACE1 [144], which is then processed by the Golgi apparatus to generate a mature BACE1. Three of the four possible N-glycosylation sites in BACE1 are used during the maturation of the molecule [144]. The protein has two open and closed conformations. The enzymatic activity reduces the open conformation by wrapping the active site in a tight conformation. BACE1 also has six cysteine residues that create three disulfide linkages. These linkages are necessary for BACE1 folding and enzymatic activity. The Cys330/Cys380 residues in BACE1’s active region are critical for their stability and activity. The activity of BACE1 is also directly proportional to its environmental pH, and a moderately acidic pH of 5.5 produces the highest activity levels. A free BACE1 also has an open flap conformation and is energetically stable, containing optimal electrovalent bonds [144].

6.3. Gamma-Secretase

Gamma-secretase cleaves CTF-beta of 99 residues, resulting in plaque-forming Aβ and CTF-gamma of 50 APP intracellular domains (AICD) plus three transmembrane domain residues [145]. Gamma secretase has an aspartyl protease activity comparable to that of bacteria’s type-4 prepilin enzyme [146], and comparable inhibitors of serine, cysteine, and aspartyl proteases can block gamma-secretase activity [147]. Gamma-secretase, however, lacks the conserved active site residues (D[TS]GS[SAT]) present in the catalytic site of nucleus-containing cell aspartyl proteases [148]. After mutation of PS1 and PS2, production of gamma-secretase increases, resulting in the development of the Aβ in AD [149]. Furthermore, the two subunits, nicastrin and anterior-pharynx defective-1, function as cofactors, forming a stable and active complex of four secretase enzymes with PS1 and PS2 [150]. Several miRNAs also boost the activity of the gamma-secretase, which results in production of Aβ; among these, MiR-9 is involved in neuronal health and synthesis of Aβ in the brain. In neurons, secretase synthesis is reduced when such complexes are inhibited, and the amyloidogenic pathways are redirected to nonamyloidogenic pathways.

6.4. Tau

Tau is a mature neuron’s main microtubule associated protein (MAP). The other two neuronal MAPs are MAP1 and MAP2. Interaction of MAPs with tubulin promotes its assembly into microtubules and increases the stability of the microtubule network, a well-established function of MAPs. Tau, a phosphoprotein, regulates its phosphorylation to promote microtubule assembly. In a normal adult human brain, tau protein has 2–3 moles of phosphate per mole. Tau hyperphosphorylation reduces tau’s biological activity. In AD, brain, tau is three to four times more hyperphosphorylated than in normal adult brain, and it polymerizes into paired helical filaments (PHF) admixed with straight filaments (SF), creating neurofibrillary tangles. Tau is transiently hyperphosphorylated throughout brain development, as well as during anesthesia and hypothermia, although not to the same extent as in AD brain samples. Tau has 352–441 amino acids and three domains (projection, proline-rich, and assembly) [151]. Extensive data imply that tau hyperphosphorylation is caused by a disruption in cellular signaling, specifically an imbalance in the activity of several protein kinases and phosphatases. It indicates that the Aβ plays a critical role in causing this imbalance in AD [152]. The active-site residues of the Tau protein (PDB ID: 6PXR) (Arg57, Ser52, Ser54, Ser53, Trp100, Asp101, and Glu39) participate in complex formation with its ligands, and this information may help in developing a tau-targeted therapeutics for AD (https://www.rcsb.org/structure/6PXR, accessed on 5 September 2022) [153].

6.5. APP Forms in AD

Chromosomes 1, 14, 11, 19, and 21 have the senilin, SORL1, APOE4, and APP genes responsible for depositing amyloid plaques and disassembling microtubules [154,155,156]. Over 32 different APP missense versions have been identified from 85 AD contracted families. Most of these mutations are located at the secretase cleavage sites or the APP transmembrane domain on exons 16 and 17. APP was first cloned by screening a cDNA library, encoding a 695 amino-acid protein consisting of 18 exons. The APP gene is located on chromosome 21q21, which can be alternatively spliced into several products, listed as APP695, 714, 751, 770, and 563. The expressed form of APP varies differently according to its tissue type, with three isoforms being most relevant to AD. The APP forms, APP 751 and APP 770, are expressed in both the peripheral and the central nervous systems [157,158].

6.6. Drugs Approved for the Treatment of AD

Studies of autosomal dominant familial AD provide the greatest evidence for the “amyloid cascade hypothesis”, a primary pathophysiologic paradigm in AD [159]. FDA-approved AD therapeutic, aducanumab, an Aβ-directed monoclonal antibody of immunoglobulin gamma 1 (IgG1) subtype, passes the blood–brain barrier (BBB) and targets the soluble oligomers and insoluble fibrils of Aβ plaques [160,161,162]. It binds to the linear epitope of the N-terminal of Aβ amino acids 3–7 [163], reduces Aβ plaques in the brain, and decreases the levels of phosphorylated tau in the CSF and medial temporal NFTs [164]. Aducanumab is given via an intravenous (IV) infusion to the patient [162]. In the last few years, various classes of inhibitors developed for beta- and gamma-secretase have reduced the accumulation of Aβ. Various compounds for beta- and gamma-secretase inhibitors have been synthesized and studied as therapeutic targets for AD. Measurements of their IC50, EC50, and Ki values (µM) are listed in Table 3 and Table 4, respectively. Compounds were synthesized by modifying the N- and C-terminal ends of beta- or gamma-secretase, and studies are ongoing for their further development. It is expected that effective candidates for AD will be reported in the future.

6.7. Other Therapeutic Approaches

Evidence suggests that Alzheimer’s disease is closely related to type 2 diabetes. Several studies in animal models highlighted the fact that insulin plays a major role in controlling various activities in the brain. Insulin enhances the clearance of Aβ and phosphorylates the tau protein. Glucagon-like peptide-1 has shown promising results in animal models by enhancing neuroregulation. Thus, it can be employed for treating Alzheimer’s disease [193]. Moreover, the inhibition of the production of Aβ by some enzymatic treatments is also a potential therapeutic approach in treating Alzheimer’s disease. The APP is cleaved by the γ-secretase and β-secretase enzymes that produce Aβ, which accumulates in the form of plagues and can cause Alzheimer’s disease [194]. Yu et al. stated that anti-neuroinflammatory therapy is a very effective treatment against AD. The astrocyte modulators, microbiome therapy, microglia modulator, and insulin resistance are four key strategies that comprise this anti-neuroinflammatory therapy [195]. The gut microbiome’s communication with the brain in both directions of the brain–microbiome–gut axis has received great attention with regard to the treatment of the Alzheimer’s disease. Therefore, the maintenance of GM, particularly the addition of probiotics, is a promising candidate for treating AD [196]. Krüger et al. performed a study with 161 subjects with AD and concluded that three randomized clinical trials patients who received Bifidobacterium and Lactobacillus strains had no benefits of probiotic supplementation on the cognitive functions. Probiotic supplementation did, however, improve insulin resistance, as well as very-low-density lipoprotein (VLDL) cholesterol, plasma triglyceride, and malondialdehyde levels. No RCTs evaluated the composition of the microbiota or incorporated synbiotic supplementation [197].

7. Microorganism-Mediated Gut Barrier Dysfunction, BBB Passage, and Activation of Chemokines/Cytokines, Leading to Breakdown of APP

7.1. Gut Inflammation and Dysbiosis

In the case of mucosal structure disruption, intestinal inflammatory processes drive polymorphonuclear cells to migrate to the gut mucosa from circulation or even farther to the gut lumen [198]. Calprotectin content in the feces can be used to quantify the process of intestinal inflammation indirectly. This tiny protein, an S100A8/A9 heterodimer, makes up about 60% of the neutrophil cytosol protein content and possesses antibacterial properties [199]. The S100A8 and S100A9 residues also contain amyloidogenic amino-acid sequences and may form amyloid oligomers and fibrils that mimic amyloid polypeptides such as Aβ and α-syn. Monomeric and dimeric S100A9 have been shown to promote Aβ fibrillization in vitro [200,201]. In one study, higher fecal calprotectin levels were discovered in nearly 70% of AD patients, and it is thought that calprotectin may translocate into circulation and contribute to neuroinflammation [202]. Such an intestinal supply of calprotectin might help promote amyloid fibril formation in the gut or directly in the brain [203].
Gut inflammation and dysbiosis are linked to gut barrier malfunction and increased intestinal permeability (termed a “leaky gut”). These may contribute to the neurodegenerative process [4]. The intestinal barrier is made up of the mucus layer, the intestinal epithelium, and the lamina propria [203]. When this barrier is breached, permeability increases, allowing germs and toxic chemicals to enter the circulation (a process called atopobiosis) [204,205,206]. The presence of mucin-degrading bacteria enhances gut barrier function and decreases obesity and systemic inflammation [204,207]. Lactobacillus plantarum, E. coli strain Nissle, and Bifidobacterium infantis are probiotic bacteria that improve the intestinal barrier by boosting the production of proteins that create tight junctions [208]. However, exotoxins, another type of bacterial substance, impair epithelial cell integrity. Changes in tight junctions have been documented for different pathogenic E. coli strains, Salmonella, Shigella, Helicobacter pylori, Vibrio, or Clostridium. For Bacteroides fragilis, its exotoxin damages adherence junctions by cleaving the cell adhesion molecules, the E-cadherin [208].
Several decades of research have also demonstrated a microbiota–gut–brain axis [209] and the alteration of APP causing AD [203] and several other neurological disorders. Fungal, viral, and bacterial microorganisms bind and activate certain surface proteins on target cells; for example, some viruses bind to the C-type lectin domain family 5 member A (CLEC5A) of macrophages and activate a cytokine and chemokine storm [144]. These bioactive chemicals are specialized to break down the tight junctions present between the endothelial cells of the BBB [210]; this allows microorganisms to enter the CNS via a transcellular route through the endothelial cells [211], causing AD. IL-6, IL-7, and TNF-α are among the known inflammatory chemicals secreted by the immune cells near the BBB (Figure 4).
Alterations in the gut microbiome have been reported to affect the etiology of AD involving immunological, endocrine, and metabolic functions. Xi J. et al. suggested that bacteria-influenced or -secreted chemicals may trigger systemic inflammatory responses known as secondary metabolites and disrupt the BBB, thus promoting neurodegeneration [212]. Oral bacteria such as P. gingivalis, Treponema, and Candida metabolites include short-chain fatty acids (SCFAs). Such SCFAs generated by local bacteria protect Th17 cells and their function in the oral mucosa in combating Candida infection. However, abnormally low or high amounts of SCFA can be a sign of inflammation or dysbiosis [213]. Additionally, it has been demonstrated in several studies that SCFA prevents Aβ aggregation [214].

7.2. Amyloid Cascade Hypothesis

For the past 20 years, research has been dominated by the amyloid cascade theory, with the premise of the buildup of the amyloid-peptide in the brain as the key event in the pathophysiology of AD. Aducanumab (Biogen) is the first FDA-approved monoclonal antibody for treating AD by removing amyloid plaques [215]. Aducanumab targets Aβ and reduces the Aβ plaques in the brains of transgenic mice [160]. The clinical trials demonstrated that the brain significantly reduced Aβ plaques in a time- and dose-dependent manner. However, increased amyloid-related imaging abnormality (ARIA) was observed depending on the dose and the APOE4 genotype (Clinical Trials: NCT01397539, NCT01677572) [216]. Studies on the effectiveness and safety of aducanumab are still not enough as FDA approved it in a hurry. Therefore, aducanumab treatment will require further validation of its continued use and instructions for a prescription. Other targeted treatments based on this hypothesis are in the research phase or being tested in clinical trials. However, some treatments intended to decrease amyloid production or aggregation have failed in clinical trials. Aβ immunotherapy with bapineuzumab, tramiprosate, and solanezumab have shown failure in phase III clinical trials, as they displayed no clinical improvements for various AD biomarkers. Similarly, in phase II clinical trials, tau aggregation inhibitors and gamma-secretase inhibitors have not shown any sufficient improvements for the various markers of AD [217,218,219,220,221,222]. Therefore, it may be important to review the science underlying the amyloid cascade hypothesis on whether amyloid-directed therapeutics will prove to be lifesaving against this debilitating disease.

8. Conclusions

AD is linked to neurological damage and progressive synaptic dysfunction. Two major proteins, the Aβ peptide and tau, have been linked to AD and, thus, used as diagnostic markers [223,224]. Chronic AD can occur due to microorganisms, which can cause the disease after being dormant in the body. Such bacterial, fungal, and viral microorganisms may contribute to AD as they become more active with a decline in systemic immunity due to aging [51]. Periodontal pathogens and gut microorganisms such as Aggregatibacter actinomycetemcomitan (Actinobacillus actinomycetemcomitans), Tannerella forsythensis, P. gingivalis, T. denticola, and Helicobacter pylori, instigate inflammation and neurotoxicity by upregulating tau phosphorylation [28]. Lactobacillus species, Bifidobacteria (Actinobacteria), Verrucomicrobia, Spirochetes, Proteobacteria, Fusobacteria, Firmicutes, and Cyanobacteria, B. burgdorferi, Chlamydia pneumoniae, E. coli, Shighella, E. rectale, and Bacteriodesfragilis have all been linked to AD. Viruses, particularly HSV, HHV, and Cytomegalovirus, facilitate neurodegeneration by boosting tau phosphorylation, accumulation of Aβ peptide, senile plaques, and neurofibrillary tangles [10,54,225]. Several reports suggest the importance of microorganisms in the pathology of AD. However, since some microorganisms are also detected in healthy individuals, projecting microorganisms as a marker for AD requires detailed studies on the association and standardization between microorganisms and AD [214,226].
There are currently few therapies available for AD, which develops from dementia, and there is no known cure. The varied drug use amongst individuals is likely the most common limitation when employing bacterial quantification from the gut in AD patients. People with Alzheimer’s and dementia must typically take medications to treat the disease’s wide range of symptoms, which can significantly impact gut composition. A person may have only a specific number of particular taxa in their microbiota, which is a significant restriction in the research of the gut microbiota of AD patients. Taken together, increasing knowledge about the association of microorganisms with AD shows that they might be projected as a potential biomarker for AD in the near future.

Author Contributions

Conceptualization, A.K.J., S.-S.L., P.Y. and Y.-H.L.; writing—original draft preparation, A.K.J., S.-S.L., P.Y. and Y.-H.L.; data curation, P.Y., Y.-H.L. and P.N.; validation, H.P., S.K., N.B., R.P., S.K.J., P.N. and N.K.J.; visualization, H.P., S.K., N.B., R.P., S.K.J. and N.K.J.; writing—review and editing, A.K.J. and S.-S.L.; supervision, A.K.J. and S.-S.L.; project administration, A.K.J. and S.-S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Hallym University Research Fund and by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1I1A3074575).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are thankful to Sharda University, Greater Noida Senior Management for constant support and encouragement.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferry, C.; Prince, M.; Brayne, C. Alzheimer’s Disease International Global prevalence of dementia: A Delphi concensus Study. Lancet 2005, 366, 2112–2117. [Google Scholar] [CrossRef]
  2. Kumar, A.; Sidhu, J.; Goyal, A.; Tsao, J.W. Alzheimer Disease; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  3. Kamer, A.R.; Craig, R.G.; Dasanayake, A.P.; Brys, M.; Glodzik-Sobanska, L.; de Leon, M.J. Inflammation and Alzheimer’s disease: Possible role of periodontal diseases. Alzheimers Dement. 2008, 4, 242–250. [Google Scholar] [CrossRef]
  4. Sochocka, M.; Donskow-Lysoniewska, K.; Diniz, B.S.; Kurpas, D.; Brzozowska, E.; Leszek, J. The Gut Microbiome Alterations and Inflammation-Driven Pathogenesis of Alzheimer’s Disease—A Critical Review. Mol. Neurobiol. 2019, 56, 1841–1851. [Google Scholar] [CrossRef] [PubMed]
  5. Landry, R.L.; Embers, M.E. Does Dementia Have a Microbial Cause? NeuroSci 2022, 3, 262–283. [Google Scholar] [CrossRef]
  6. 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]
  7. Wozniak, M.A.; Mee, A.P.; Itzhaki, R.F. Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaques. J. Pathol. 2009, 217, 131–138. [Google Scholar] [CrossRef]
  8. Miklossy, J. Alzheimer’s disease--a spirochetosis? Neuroreport 1993, 4, 841–848. [Google Scholar] [CrossRef]
  9. Miklossy, J.; Kis, A.; Radenovic, A.; Miller, L.; Forro, L.; Martins, R.; Reiss, K.; Darbinian, N.; Darekar, P.; Mihaly, L.; et al. Beta-amyloid deposition and Alzheimer’s type changes induced by Borrelia spirochetes. Neurobiol. Aging 2006, 27, 228–236. [Google Scholar] [CrossRef] [PubMed]
  10. Wozniak, M.A.; Itzhaki, R.F.; Shipley, S.J.; Dobson, C.B. Herpes simplex virus infection causes cellular beta-amyloid accumulation and secretase upregulation. Neurosci. Lett. 2007, 429, 95–100. [Google Scholar] [CrossRef]
  11. Ramesh, G.; Alvarez, A.L.; Roberts, E.D.; Dennis, V.A.; Lasater, B.L.; Alvarez, X.; Philipp, M.T. Pathogenesis of Lyme neuroborreliosis: Borrelia burgdorferi lipoproteins induce both proliferation and apoptosis in rhesus monkey astrocytes. Eur. J. Immunol. 2003, 33, 2539–2550. [Google Scholar] [CrossRef] [PubMed]
  12. Ho, G.J.; Drego, R.; Hakimian, E.; Masliah, E. Mechanisms of cell signaling and inflammation in Alzheimer’s disease. Curr. Drug Targets Inflamm. Allergy 2005, 4, 247–256. [Google Scholar] [CrossRef]
  13. Wyss-Coray, T.; Rogers, J. Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harb. Perspect Med. 2012, 2, a006346. [Google Scholar] [CrossRef]
  14. Dosunmu, R.; Wu, J.; Basha, M.R.; Zawia, N.H. Environmental and dietary risk factors in Alzheimer’s disease. Expert Rev. Neurother. 2007, 7, 887–900. [Google Scholar] [CrossRef] [PubMed]
  15. Bulgart, H.R.; Neczypor, E.W.; Wold, L.E.; Mackos, A.R. Microbial involvement in Alzheimer disease development and progression. Mol. Neurodegener. 2020, 15, 42. [Google Scholar] [CrossRef] [PubMed]
  16. Askarova, S.; Umbayev, B.; Masoud, A.R.; Kaiyrlykyzy, A.; Safarova, Y.; Tsoy, A.; Olzhayev, F.; Kushugulova, A. The Links Between the Gut Microbiome, Aging, Modern Lifestyle and Alzheimer’s Disease. Front. Cell Infect. Microbiol. 2020, 10, 104. [Google Scholar] [CrossRef] [PubMed]
  17. Rajmohan, R.; Reddy, P.H. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer’s disease Neurons. J. Alzheimers Dis. 2017, 57, 975–999. [Google Scholar] [CrossRef]
  18. Murphy, M.P.; LeVine, H., 3rd. Alzheimer’s disease and the amyloid-beta peptide. J. Alzheimers Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef] [PubMed]
  19. Sun, X.; Chen, W.D.; Wang, Y.D. beta-Amyloid: The key peptide in the pathogenesis of Alzheimer’s disease. Front. Pharmacol. 2015, 6, 221. [Google Scholar] [CrossRef]
  20. Guo, J.; Jia, X.; Liu, Y.; Wang, S.; Cao, J.; Zhang, B.; Xiao, G.; Wang, W. Screening of Natural Extracts for Inhibitors against Japanese Encephalitis Virus Infection. Antimicrob. Agents Chemother. 2020, 64, e02373-19. [Google Scholar] [CrossRef] [PubMed]
  21. Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lim, J.; Yue, Z. Neuronal aggregates: Formation, clearance, and spreading. Dev. Cell 2015, 32, 491–501. [Google Scholar] [CrossRef]
  23. Wang, Y.C.; Lauwers, E.; Verstreken, P. Presynaptic protein homeostasis and neuronal function. Curr. Opin. Genet. Dev. 2017, 44, 38–46. [Google Scholar] [CrossRef] [PubMed]
  24. De Wilde, M.C.; Overk, C.R.; Sijben, J.W.; Masliah, E. Meta-analysis of synaptic pathology in Alzheimer’s disease reveals selective molecular vesicular machinery vulnerability. Alzheimers Dement. 2016, 12, 633–644. [Google Scholar] [CrossRef] [PubMed]
  25. Spires-Jones, T.L.; Hyman, B.T. The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron 2014, 82, 756–771. [Google Scholar] [CrossRef]
  26. Herrera-Landero, A.; Amaya-Sanchez, L.E.; Solorzano-Santos, F.; Gordillo-Perez, M.G. Borrelia burgdorferi as a risk factor for Alzheimer’s dementia and mild cognitive impairment. Eur. Geriatr. Med. 2019, 10, 493–500. [Google Scholar] [CrossRef] [PubMed]
  27. Senejani, A.G.; Maghsoudlou, J.; El-Zohiry, D.; Gaur, G.; Wawrzeniak, K.; Caravaglia, C.; Khatri, V.A.; MacDonald, A.; Sapi, E. Borrelia burgdorferi Co-Localizing with Amyloid Markers in Alzheimer’s Disease Brain Tissues. J. Alzheimers Dis. 2022, 85, 889–903. [Google Scholar] [CrossRef] [PubMed]
  28. Haahr, R.; Tetens, M.M.; Dessau, R.B.; Krogfelt, K.A.; Bodilsen, J.; Andersen, N.S.; Moller, J.K.; Roed, C.; Christiansen, C.B.; Ellermann-Eriksen, S.; et al. Risk of Neurological Disorders in Patients With European Lyme Neuroborreliosis: A Nationwide, Population-Based Cohort Study. Clin. Infect. Dis. 2020, 71, 1511–1516. [Google Scholar] [CrossRef]
  29. Ramesh, G.; Borda, J.T.; Dufour, J.; Kaushal, D.; Ramamoorthy, R.; Lackner, A.A.; Philipp, M.T. Interaction of the Lyme disease spirochete Borrelia burgdorferi with brain parenchyma elicits inflammatory mediators from glial cells as well as glial and neuronal apoptosis. Am. J. Pathol. 2008, 173, 1415–1427. [Google Scholar] [CrossRef] [PubMed]
  30. Poole, S.; Singhrao, S.K.; Kesavalu, L.; Curtis, M.A.; Crean, S. Determining the presence of periodontopathic virulence factors in short-term postmortem Alzheimer’s disease brain tissue. J. Alzheimers Dis. 2013, 36, 665–677. [Google Scholar] [CrossRef]
  31. Díaz-Zúñiga, J.; Muñoz, Y.; Melgar-Rodríguez, S.; More, J.; Bruna, B.; Lobos, P.; Monasterio, G.; Vernal, R.; Paula-Lima, A. Serotype b of Aggregatibacter actinomycetemcomitans triggers pro-inflammatory responses and amyloid beta secretion in hippocampal cells: A novel link between periodontitis and Alzheimer’s disease? J. Oral Microbiol. 2019, 11, 1586423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ellen, R.P.; Galimanas, V.B. Spirochetes at the forefront of periodontal infections. Periodontology 2000 2005, 38, 13–32. [Google Scholar] [CrossRef] [PubMed]
  33. Dioguardi, M.; Crincoli, V.; Laino, L.; Alovisi, M.; Sovereto, D.; Mastrangelo, F.; Lo Russo, L.; Lo Muzio, L. The role of periodontitis and periodontal bacteria in the onset and progression of Alzheimer’s disease: A systematic review. J. Clin. Med. 2020, 9, 495. [Google Scholar] [CrossRef] [PubMed]
  34. Miklossy, J. Historic evidence to support a causal relationship between spirochetal infections and Alzheimer’s disease. Front. Aging Neurosci. 2015, 7, 46. [Google Scholar] [CrossRef] [PubMed]
  35. Foschi, F.; Izard, J.; Sasaki, H.; Sambri, V.; Prati, C.; Müller, R.; Stashenko, P. Treponema denticola in disseminating endodontic infections. J. Dent. Res. 2006, 85, 761–765. [Google Scholar] [CrossRef]
  36. Riviere, G.R.; Riviere, K.H.; Smith, K.S. Molecular and immunological evidence of oral Treponema in the human brain and their association with Alzheimer’s disease. Oral. Microbiol. Immunol. 2002, 17, 113–118. [Google Scholar] [CrossRef]
  37. Zhuang, Z.Q.; Shen, L.L.; Li, W.W.; Fu, X.; Zeng, F.; Gui, L.; Lu, Y.; Cai, M.; Zhu, C.; Tan, Y.L.; et al. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2018, 63, 1337–1346. [Google Scholar] [CrossRef]
  38. Henderson, G.; Jaber, T.; Carpenter, D.; Wechsler, S.L.; Jones, C. Identification of herpes simplex virus type 1 proteins encoded within the first 1.5 kb of the latency-associated transcript. J. Neurovirol. 2009, 15, 439–448. [Google Scholar] [CrossRef]
  39. Porcellini, E.; Carbone, I.; Ianni, M.; Licastro, F. Alzheimer’s disease gene signature says: Beware of brain viral infections. Immun. Ageing 2010, 7, 16. [Google Scholar] [CrossRef]
  40. Wozniak, M.; Bell, T.; Denes, A.; Falshaw, R.; Itzhaki, R. Anti-HSV1 activity of brown algal polysaccharides and possible relevance to the treatment of Alzheimer’s disease. Int. J. Biol. Macromol. 2015, 74, 530–540. [Google Scholar] [CrossRef]
  41. Westman, G.; Blomberg, J.; Yun, Z.; Lannfelt, L.; Ingelsson, M.; Eriksson, B.M. Decreased HHV-6 IgG in Alzheimer’s Disease. Front. Neurol. 2017, 8, 40. [Google Scholar] [CrossRef] [Green Version]
  42. Itzhaki, R.F. Herpes simplex virus type 1 and Alzheimer’s disease: Possible mechanisms and signposts. FASEB J. 2017, 31, 3216–3226. [Google Scholar] [CrossRef] [PubMed]
  43. De Chiara, G.; Marcocci, M.E.; Sgarbanti, R.; Civitelli, L.; Ripoli, C.; Piacentini, R.; Garaci, E.; Grassi, C.; Palamara, A.T. Infectious agents and neurodegeneration. Mol. Neurobiol. 2012, 46, 614–638. [Google Scholar] [CrossRef] [PubMed]
  44. Hill, J.M.; Clement, C.; Pogue, A.I.; Bhattacharjee, S.; Zhao, Y.; Lukiw, W.J. Pathogenic microbes, the microbiome, and Alzheimer’s disease (AD). Front. Aging Neurosci. 2014, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  45. Cairns, D.M.; Rouleau, N.; Parker, R.N.; Walsh, K.G.; Gehrke, L.; Kaplan, D.L. A 3D human brain-like tissue model of herpes-induced Alzheimer’s disease. Sci. Adv. 2020, 6, eaay8828. [Google Scholar] [CrossRef] [PubMed]
  46. Bradshaw, M.J.; Venkatesan, A. Herpes simplex virus-1 encephalitis in adults: Pathophysiology, diagnosis, and management. Neurotherapeutics 2016, 13, 493–508. [Google Scholar] [CrossRef] [PubMed]
  47. Herpes Simplex Virus. World Health Organization (WHO). Available online: https://www.who.int/news-room/fact-sheets/detail/herpes-simplex-virus (accessed on 3 July 2022).
  48. Protto, V.; Marcocci, M.E.; Miteva, M.T.; Piacentini, R.; Li Puma, D.D.; Grassi, C.; Palamara, A.T.; De Chiara, G. Role of HSV-1 in Alzheimer’s disease pathogenesis: A challenge for novel preventive/therapeutic strategies. Curr. Opin. Pharmacol. 2022, 63, 102200. [Google Scholar] [CrossRef]
  49. Harris, S.A.; Harris, E.A. Herpes simplex virus type 1 and other pathogens are key causative factors in sporadic Alzheimer’s disease. J. Alzheimers Dis. 2015, 48, 319–353. [Google Scholar] [CrossRef]
  50. Itzhaki, R.F.; Lin, W.R.; Shang, D.; Wilcock, G.K.; Faragher, B.; Jamieson, G.A. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet 1997, 349, 241–244. [Google Scholar] [CrossRef]
  51. Itzhaki, R.F. Herpes simplex virus type 1 and Alzheimer’s disease: Increasing evidence for a major role of the virus. Front. Aging Neurosci. 2014, 6, 202. [Google Scholar] [CrossRef]
  52. Mawanda, F.; Wallace, R. Can infections cause Alzheimer’s disease? Epidemiol. Rev. 2013, 35, 161–180. [Google Scholar] [CrossRef] [Green Version]
  53. Carter, C. Interactions between the products of the Herpes simplex genome and Alzheimer’s disease susceptibility genes: Relevance to pathological-signalling cascades. Neurochem. Int. 2008, 52, 920–934. [Google Scholar] [CrossRef] [PubMed]
  54. Harris, S.A.; Harris, E.A. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer’s Disease. Front. Aging Neurosci. 2018, 10, 48. [Google Scholar] [CrossRef] [PubMed]
  55. Campadelli-Fiume, G.; Mirandola, P.; Menotti, L. Human herpesvirus 6: An emerging pathogen. Emerg. Infect. Dis. 1999, 5, 353–366. [Google Scholar] [CrossRef]
  56. Readhead, B.; Haure-Mirande, J.V.; Funk, C.C.; Richards, M.A.; Shannon, P.; Haroutunian, V.; Sano, M.; Liang, W.S.; Beckmann, N.D.; Price, N.D.; et al. Multiscale Analysis of Independent Alzheimer’s Cohorts Finds Disruption of Molecular, Genetic, and Clinical Networks by Human Herpesvirus. Neuron 2018, 99, 64–82.e67. [Google Scholar] [CrossRef] [PubMed]
  57. Alonso, R.; Pisa, D.; Fernandez-Fernandez, A.M.; Carrasco, L. Infection of Fungi and Bacteria in Brain Tissue From Elderly Persons and Patients With Alzheimer’s Disease. Front. Aging Neurosci. 2018, 10, 159. [Google Scholar] [CrossRef] [PubMed]
  58. Alonso, R.; Pisa, D.; Marina, A.I.; Morato, E.; Rabano, A.; Carrasco, L. Fungal infection in patients with Alzheimer’s disease. J. Alzheimers Dis. 2014, 41, 301–311. [Google Scholar] [CrossRef] [PubMed]
  59. Alonso, R.; Pisa, D.; Rábano, A.; Carrasco, L. Alzheimer’s disease and disseminated mycoses. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1125–1132. [Google Scholar] [CrossRef]
  60. Perez, J.C. The interplay between gut bacteria and the yeast Candida albicans. Gut Microbes 2021, 13, 1979877. [Google Scholar] [CrossRef]
  61. Garcia, R.I.; Henshaw, M.M.; Krall, E.A. Relationship between periodontal disease and systemic health. Periodontology 2000 2001, 25, 21–36. [Google Scholar] [CrossRef]
  62. Abbayya, K.; Puthanakar, N.Y.; Naduwinmani, S.; Chidambar, Y. Association between periodontitis and Alzheimer’s disease. N. Am. J. Med. Sci. 2015, 7, 241. [Google Scholar] [CrossRef] [Green Version]
  63. Tonsekar, P.P.; Jiang, S.S.; Yue, G. Periodontal disease, tooth loss and dementia: Is there a link? A systematic review. Gerodontology 2017, 34, 151–163. [Google Scholar] [CrossRef]
  64. Socransky, S.S.; Haffajee, A.D. Periodontal microbial ecology. Periodontology 2000 2005, 38, 135–187. [Google Scholar] [CrossRef] [PubMed]
  65. Kamer, A.R.; Fortea, J.O.; Videla, S.; Mayoral, A.; Janal, M.; Carmona-Iragui, M.; Benejam, B.; Craig, R.G.; Saxena, D.; Corby, P.; et al. Periodontal disease’s contribution to Alzheimer’s disease progression in Down syndrome. Alzheimers Dement. 2016, 2, 49–57. [Google Scholar] [CrossRef]
  66. 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]
  67. Varesi, A.; Pierella, E.; Romeo, M.; Piccini, G.B.; Alfano, C.; Bjorklund, G.; Oppong, A.; Ricevuti, G.; Esposito, C.; Chirumbolo, S.; et al. The Potential Role of Gut Microbiota in Alzheimer’s Disease: From Diagnosis to Treatment. Nutrients 2022, 14, 668. [Google Scholar] [CrossRef] [PubMed]
  68. Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef]
  69. Sankowski, R.; Mader, S.; Valdes-Ferrer, S.I. Systemic inflammation and the brain: Novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front. Cell Neurosci. 2015, 9, 28. [Google Scholar] [CrossRef] [PubMed]
  70. Zhao, J.; Bi, W.; Xiao, S.; Lan, X.; Cheng, X.; Zhang, J.; Lu, D.; Wei, W.; Wang, Y.; Li, H.; et al. Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci. Rep. 2019, 9, 5790. [Google Scholar] [CrossRef] [PubMed]
  71. Gutsmann, T.; Muller, M.; Carroll, S.F.; MacKenzie, R.C.; Wiese, A.; Seydel, U. Dual role of lipopolysaccharide (LPS)-binding protein in neutralization of LPS and enhancement of LPS-induced activation of mononuclear cells. Infect. Immun. 2001, 69, 6942–6950. [Google Scholar] [CrossRef] [PubMed]
  72. Tsukamoto, H.; Takeuchi, S.; Kubota, K.; Kobayashi, Y.; Kozakai, S.; Ukai, I.; Shichiku, A.; Okubo, M.; Numasaki, M.; Kanemitsu, Y.; et al. Lipopolysaccharide (LPS)-binding protein stimulates CD14-dependent Toll-like receptor 4 internalization and LPS-induced TBK1-IKK-IRF3 axis activation. J. Biol. Chem. 2018, 293, 10186–10201. [Google Scholar] [CrossRef] [Green Version]
  73. Balin, B.J.; Gérard, H.C.; Arking, E.J.; Appelt, D.M.; Branigan, P.J.; Abrams, J.T.; Whittum-Hudson, J.A.; Hudson, A.P. Identification and localization of Chlamydia pneumoniae in the Alzheimer’s brain. Med. Microbiol. Immunol. 1998, 187, 23–42. [Google Scholar] [CrossRef]
  74. Burns, A.; Jacoby, R.; Luthert, P.; Levy, R. Cause of death in Alzheimer’s disease. Age Ageing 1990, 19, 341–344. [Google Scholar] [CrossRef]
  75. Liu, G.; Lanham, C.; Buchan, J.R.; Kaplan, M.E. High-throughput transformation of Saccharomyces cerevisiae using liquid handling robots. PLoS ONE 2017, 12, e0174128. [Google Scholar] [CrossRef]
  76. Dhakal, S.; Macreadie, I. Protein homeostasis networks and the use of yeast to guide interventions in Alzheimer’s disease. Int. J. Mol. Sci. 2020, 21, 8014. [Google Scholar] [CrossRef] [PubMed]
  77. Bharadwaj, P.; Martins, R.; Macreadie, I. Yeast as a model for studying Alzheimer’s disease. FEMS Yeast Res. 2010, 10, 961–969. [Google Scholar] [CrossRef]
  78. Rencus-Lazar, S.; DeRowe, Y.; Adsi, H.; Gazit, E.; Laor, D. Yeast Models for the Study of Amyloid-Associated Disorders and Development of Future Therapy. Front. Mol. Biosci. 2019, 6, 15. [Google Scholar] [CrossRef]
  79. Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 1–10. [Google Scholar] [CrossRef]
  80. Seo, D.O.; Holtzman, D.M. Gut Microbiota: From the Forgotten Organ to a Potential Key Player in the Pathology of Alzheimer’s Disease. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1232–1241. [Google Scholar] [CrossRef] [PubMed]
  81. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef]
  82. Piacentini, E.; Sanchez, B.; Arauzo, V.; Calbo, E.; Cuchi, E.; Nava, J.M. Procalcitonin levels are lower in intensive care unit patients with H1N1 influenza A virus pneumonia than in those with community-acquired bacterial pneumonia. A pilot study. J. Crit. Care 2011, 26, 201–205. [Google Scholar] [CrossRef] [PubMed]
  83. Wu, L.; Han, Y.; Zheng, Z.; Peng, G.; Liu, P.; Yue, S.; Zhu, S.; Chen, J.; Lv, H.; Shao, L.; et al. Altered Gut Microbial Metabolites in Amnestic Mild Cognitive Impairment and Alzheimer’s Disease: Signals in Host-Microbe Interplay. Nutrients 2021, 13, 228. [Google Scholar] [CrossRef]
  84. Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef] [PubMed]
  85. Bibi, F.; Yasir, M.; Sohrab, S.S.; Azhar, E.I.; Al-Qahtani, M.H.; Abuzenadah, A.M.; Kamal, M.A.; Naseer, M.I. Link between chronic bacterial inflammation and Alzheimer disease. CNS Neurol. Disord. Drug Targets Former. Curr. Drug Targets-CNS Neurol. Disord. 2014, 13, 1140–1147. [Google Scholar] [CrossRef]
  86. Zhao, Y.; Jaber, V.; Lukiw, W.J. Secretory Products of the Human GI Tract Microbiome and Their Potential Impact on Alzheimer’s Disease (AD): Detection of Lipopolysaccharide (LPS) in AD Hippocampus. Front. Cell Infect. Microbiol. 2017, 7, 318. [Google Scholar] [CrossRef]
  87. Johnson, K.V.; Foster, K.R. Why does the microbiome affect behaviour? Nat. Rev. Microbiol. 2018, 16, 647–655. [Google Scholar] [CrossRef]
  88. Bostanciklioğlu, M. Intestinal bacterial flora and Alzheimer’s disease. Neurophysiology 2018, 50, 140–148. [Google Scholar] [CrossRef]
  89. Li, B.; He, Y.; Ma, J.; Huang, P.; Du, J.; Cao, L.; Wang, Y.; Xiao, Q.; Tang, H.; Chen, S. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019, 15, 1357–1366. [Google Scholar] [CrossRef]
  90. He, Y.; Li, B.; Sun, D.; Chen, S. Gut Microbiota: Implications in Alzheimer’s Disease. J. Clin. Med. 2020, 9, 2042. [Google Scholar] [CrossRef]
  91. Wang, Y.K.; Kuo, F.C.; Liu, C.J.; Wu, M.C.; Shih, H.Y.; Wang, S.S.; Wu, J.Y.; Kuo, C.H.; Huang, Y.K.; Wu, D.C. Diagnosis of Helicobacter pylori infection: Current options and developments. World J. Gastroenterol. 2015, 21, 11221–11235. [Google Scholar] [CrossRef] [PubMed]
  92. Bu, X.L.; Yao, X.Q.; Jiao, S.S.; Zeng, F.; Liu, Y.H.; Xiang, Y.; Liang, C.R.; Wang, Q.H.; Wang, X.; Cao, H.Y. A study on the association between infectious burden and A lzheimer’s disease. Eur. J. Neurol. 2015, 22, 1519–1525. [Google Scholar] [CrossRef] [PubMed]
  93. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef]
  94. Hudec, J.; Kobida, L.; Canigova, M.; Lacko-Bartosova, M.; Lozek, O.; Chlebo, P.; Mrazova, J.; Ducsay, L.; Bystricka, J. Production of gamma-aminobutyric acid by microorganisms from different food sources. J. Sci. Food Agric. 2015, 95, 1190–1198. [Google Scholar] [CrossRef]
  95. Mitew, S.; Kirkcaldie, M.T.; Dickson, T.C.; Vickers, J.C. Altered synapses and gliotransmission in Alzheimer’s disease and AD model mice. Neurobiol. Aging 2013, 34, 2341–2351. [Google Scholar] [CrossRef]
  96. Brown, G.C. The endotoxin hypothesis of neurodegeneration. J. Neuroinflamm. 2019, 16, 1–10. [Google Scholar] [CrossRef]
  97. Zhan, X.; Stamova, B.; Sharp, F.R. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer’s Disease Brain: A Review. Front. Aging Neurosci. 2018, 10, 42. [Google Scholar] [CrossRef]
  98. Hoogland, I.C.; Houbolt, C.; van Westerloo, D.J.; van Gool, W.A.; van de Beek, D. Systemic inflammation and microglial activation: Systematic review of animal experiments. J. Neuroinflamm. 2015, 12, 114. [Google Scholar] [CrossRef]
  99. Miklossy, J. Emerging roles of pathogens in Alzheimer disease. Expert Rev. Mol. Med. 2011, 13, e30. [Google Scholar] [CrossRef]
  100. Cheon, M.S.; Bajo, M.; Gulesserian, T.; Cairns, N.; Lubec, G. Evidence for the relation of herpes simplex virus type 1 to Down syndrome and Alzheimer’s disease. Electrophoresis 2001, 22, 445–448. [Google Scholar] [CrossRef]
  101. Lin, W.R.; Wozniak, M.A.; Cooper, R.J.; Wilcock, G.K.; Itzhaki, R.F. Herpesviruses in brain and Alzheimer’s disease. J. Pathol. 2002, 197, 395–402. [Google Scholar] [CrossRef]
  102. Strandberg, T.E.; Pitkala, K.; Eerola, J.; Tilvis, R.; Tienari, P.J. Interaction of herpesviridae, APOE gene, and education in cognitive impairment. Neurobiol. Aging 2005, 26, 1001–1004. [Google Scholar] [CrossRef]
  103. Katan, M.; Moon, Y.P.; Paik, M.C.; Sacco, R.L.; Wright, C.B.; Elkind, M.S. Infectious burden and cognitive function: The Northern Manhattan Study. Neurology 2013, 80, 1209–1215. [Google Scholar] [CrossRef]
  104. Wright, C.B.; Gardener, H.; Dong, C.; Yoshita, M.; DeCarli, C.; Sacco, R.L.; Stern, Y.; Elkind, M.S. Infectious Burden and Cognitive Decline in the Northern Manhattan Study. J. Am. Geriatr. Soc. 2015, 63, 1540–1545. [Google Scholar] [CrossRef]
  105. Emery, D.C.; Shoemark, D.K.; Batstone, T.E.; Waterfall, C.M.; Coghill, J.A.; Cerajewska, T.L.; Davies, M.; West, N.X.; Allen, S.J. 16S rRNA next generation sequencing analysis shows bacteria in Alzheimer’s post-mortem brain. Front. Aging Neurosci. 2017, 9, 195. [Google Scholar] [CrossRef] [PubMed]
  106. Alkasir, R.; Li, J.; Li, X.; Jin, M.; Zhu, B. Human gut microbiota: The links with dementia development. Protein Cell 2017, 8, 90–102. [Google Scholar] [CrossRef] [PubMed]
  107. Kamer, A.R.; Craig, R.G.; Pirraglia, E.; Dasanayake, A.P.; Norman, R.G.; Boylan, R.J.; Nehorayoff, A.; Glodzik, L.; Brys, M.; de Leon, M.J. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J. Neuroimmunol. 2009, 216, 92–97. [Google Scholar] [CrossRef]
  108. Sparks Stein, P.; Steffen, M.J.; Smith, C.; Jicha, G.; Ebersole, J.L.; Abner, E.; Dawson, D., 3rd. Serum antibodies to periodontal pathogens are a risk factor for Alzheimer’s disease. Alzheimers Dement. 2012, 8, 196–203. [Google Scholar] [CrossRef] [PubMed]
  109. Siddiqui, H.; Eribe, E.R.; Singhrao, S.K.; Olsen, I. High throughput sequencing detect gingivitis and periodontal oral bacteria in Alzheimer’s disease autopsy brains. J. Neurosci. Res. 2019, 1, 1–10. [Google Scholar] [CrossRef]
  110. Beydoun, M.A.; Beydoun, H.A.; Weiss, J.; Hossain, S.; El-Hajj, Z.W.; Zonderman, A.B. Helicobacter pylori, periodontal pathogens, and their interactive association with incident all-cause and Alzheimer’s disease dementia in a large national survey. Mol. Psychiatry 2021, 26, 6038–6053. [Google Scholar] [CrossRef]
  111. Kanagasingam, S.; Chukkapalli, S.S.; Welbury, R.; Singhrao, S.K. Porphyromonas gingivalis is a Strong Risk Factor for Alzheimer’s Disease. J. Alzheimers Dis. Rep. 2020, 4, 501–511. [Google Scholar] [CrossRef] [PubMed]
  112. Gale, S.D.; Brown, B.L.; Erickson, L.; Berrett, A.; Hedges, D.W. Association between latent toxoplasmosis and cognition in adults: A cross-sectional study. Parasitology 2015, 142, 557–565. [Google Scholar] [CrossRef] [PubMed]
  113. Lovheim, H.; Olsson, J.; Weidung, B.; Johansson, A.; Eriksson, S.; Hallmans, G.; Elgh, F. Interaction between Cytomegalovirus and Herpes Simplex Virus Type 1 Associated with the Risk of Alzheimer’s Disease Development. J. Alzheimers Dis. 2018, 61, 939–945. [Google Scholar] [CrossRef]
  114. Hemling, N.; Roytta, M.; Rinne, J.; Pollanen, P.; Broberg, E.; Tapio, V.; Vahlberg, T.; Hukkanen, V. Herpesviruses in brains in Alzheimer’s and Parkinson’s diseases. Ann. Neurol. 2003, 54, 267–271. [Google Scholar] [CrossRef]
  115. Carbone, I.; Lazzarotto, T.; Ianni, M.; Porcellini, E.; Forti, P.; Masliah, E.; Gabrielli, L.; Licastro, F. Herpes virus in Alzheimer’s disease: Relation to progression of the disease. Neurobiol. Aging 2014, 35, 122–129. [Google Scholar] [CrossRef]
  116. Alonso, R.; Pisa, D.; Aguado, B.; Carrasco, L. Identification of fungal species in brain tissue from Alzheimer’s disease by next-generation sequencing. J. Alzheimers Dis. 2017, 58, 55–67. [Google Scholar] [CrossRef]
  117. Alonso, R.; Pisa, D.; Fernández-Fernández, A.M.; Rábano, A.; Carrasco, L. Fungal infection in neural tissue of patients with amyotrophic lateral sclerosis. Neurobiol. Dis. 2017, 108, 249–260. [Google Scholar] [CrossRef]
  118. Brandscheid, C.; Schuck, F.; Reinhardt, S.; Schafer, K.H.; Pietrzik, C.U.; Grimm, M.; Hartmann, T.; Schwiertz, A.; Endres, K. Altered Gut Microbiome Composition and Tryptic Activity of the 5xFAD Alzheimer’s Mouse Model. J. Alzheimers Dis. 2017, 56, 775–788. [Google Scholar] [CrossRef]
  119. Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef] [PubMed]
  120. Singhrao, S.K.; Harding, A.; Poole, S.; Kesavalu, L.; Crean, S. Porphyromonas gingivalis Periodontal Infection and Its Putative Links with Alzheimer’s Disease. Mediat. Inflamm. 2015, 2015, 137357. [Google Scholar] [CrossRef]
  121. Minter, M.R.; Zhang, C.; Leone, V.; Ringus, D.L.; Zhang, X.; Oyler-Castrillo, P.; Musch, M.W.; Liao, F.; Ward, J.F.; Holtzman, D.M.; et al. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Sci. Rep. 2016, 6, 30028. [Google Scholar] [CrossRef] [PubMed]
  122. Minter, M.R.; Hinterleitner, R.; Meisel, M.; Zhang, C.; Leone, V.; Zhang, X.; Oyler-Castrillo, P.; Zhang, X.; Musch, M.W.; Shen, X.; et al. Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1DeltaE9 murine model of Alzheimer’s disease. Sci. Rep. 2017, 7, 10411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Harach, T.; Marungruang, N.; Duthilleul, N.; Cheatham, V.; Mc Coy, K.D.; Frisoni, G.; Neher, J.J.; Fak, F.; Jucker, M.; Lasser, T.; et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci. Rep. 2017, 7, 41802. [Google Scholar] [CrossRef]
  124. Bauerl, C.; Collado, M.C.; Diaz Cuevas, A.; Vina, J.; Perez Martinez, G. Shifts in gut microbiota composition in an APP/PSS1 transgenic mouse model of Alzheimer’s disease during lifespan. Lett. Appl. Microbiol. 2018, 66, 464–471. [Google Scholar] [CrossRef]
  125. Kobayashi, Y.; Sugahara, H.; Shimada, K.; Mitsuyama, E.; Kuhara, T.; Yasuoka, A.; Kondo, T.; Abe, K.; Xiao, J.Z. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer’s disease. Sci. Rep. 2017, 7, 13510. [Google Scholar] [CrossRef]
  126. Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, KC01–KC05. [Google Scholar] [CrossRef]
  127. Azm, S.A.N.; Djazayeri, A.; Safa, M.; Azami, K.; Ahmadvand, B.; Sabbaghziarani, F.; Sharifzadeh, M.; Vafa, M. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in beta-amyloid (1-42) injected rats. Appl. Physiol. Nutr. Metab. 2018, 43, 718–726. [Google Scholar] [CrossRef]
  128. Wu, S.C.; Cao, Z.S.; Chang, K.M.; Juang, J.L. Intestinal microbial dysbiosis aggravates the progression of Alzheimer’s disease in Drosophila. Nat. Commun. 2017, 8, 24. [Google Scholar] [CrossRef] [PubMed]
  129. Guilherme, M.D.S.; Nguyen, V.T.T.; Reinhardt, C.; Endres, K. Impact of Gut Microbiome Manipulation in 5xFAD Mice on Alzheimer’s Disease-Like Pathology. Microorganisms 2021, 9, 815. [Google Scholar] [CrossRef] [PubMed]
  130. Lu, B.; Vogel, H. Drosophila models of neurodegenerative diseases. Annu. Rev. Pathol. 2009, 4, 315–342. [Google Scholar] [CrossRef]
  131. Van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.; Tijssen, J.G.; et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef]
  132. Hazan, S. Rapid improvement in Alzheimer’s disease symptoms following fecal microbiota transplantation: A case report. J. Int. Med. Res. 2020, 48, 300060520925930. [Google Scholar] [CrossRef] [PubMed]
  133. Mancuso, C.; Santangelo, R. Alzheimer’s disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol. Res. 2018, 129, 329–336. [Google Scholar] [CrossRef]
  134. Leblhuber, F.; Egger, M.; Schuetz, B.; Fuchs, D. Commentary: Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2018, 10, 54. [Google Scholar] [CrossRef]
  135. Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The Gut Microbiota and Alzheimer’s Disease. J. Alzheimers Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef]
  136. Zhan, G.; Yang, N.; Li, S.; Huang, N.; Fang, X.; Zhang, J.; Zhu, B.; Yang, L.; Yang, C.; Luo, A. Abnormal gut microbiota composition contributes to cognitive dysfunction in SAMP8 mice. Aging 2018, 10, 1257–1267. [Google Scholar] [CrossRef]
  137. Vassar, R.; Kandalepas, P.C. The beta-secretase enzyme BACE1 as a therapeutic target for Alzheimer’s disease. Alzheimers Res. Ther. 2011, 3, 20. [Google Scholar] [CrossRef]
  138. Hampel, H.; Vassar, R.; De Strooper, B.; Hardy, J.; Willem, M.; Singh, N.; Zhou, J.; Yan, R.; Vanmechelen, E.; De Vos, A. The β-secretase BACE1 in Alzheimer’s disease. Biol. Psychiatry 2021, 89, 745–756. [Google Scholar] [CrossRef]
  139. Yang, G.; Zhou, R.; Guo, X.; Yan, C.; Lei, J.; Shi, Y. Structural basis of gamma-secretase inhibition and modulation by small molecule drugs. Cell 2021, 184, 521–533.e514. [Google Scholar] [CrossRef]
  140. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef]
  141. Zhang, X.; Song, W. The role of APP and BACE1 trafficking in APP processing and amyloid-beta generation. Alzheimers Res. Ther. 2013, 5, 46. [Google Scholar] [CrossRef]
  142. Cole, S.L.; Vassar, R. The Alzheimer’s disease β-secretase enzyme, BACE1. Mol. Neurodegener. 2007, 2, 1–25. [Google Scholar] [CrossRef] [Green Version]
  143. Venugopal, C.; Demos, C.M.; Rao, K.S.; Pappolla, M.A.; Sambamurti, K. Beta-secretase: Structure, function, and evolution. CNS Neurol. Disord. Drug Targets 2008, 7, 278–294. [Google Scholar] [CrossRef]
  144. Ghosh, A.K.; Osswald, H.L. BACE1 (β-secretase) inhibitors for the treatment of Alzheimer’s disease. Chem. Soc. Rev. 2014, 43, 6765–6813. [Google Scholar] [CrossRef]
  145. Chu, J.; Li, J.G.; Hoffman, N.E.; Madesh, M.; Praticò, D. Degradation of gamma secretase activating protein by the ubiquitin–proteasome pathway. J. Neurochem. 2015, 133, 432–439. [Google Scholar] [CrossRef]
  146. Wolfe, M.S. gamma-Secretase as a target for Alzheimer’s disease. Adv. Pharmacol. 2012, 64, 127–153. [Google Scholar] [CrossRef]
  147. Gertsik, N.; Chiu, D.; LI, Y. Complex regulation of gamma-secretase: From obligatory to modulatory subunits. Front. Aging Neurosci. 2015, 6, 342. [Google Scholar] [CrossRef]
  148. Haugabook, S.; Sambamurti, K. Gamma Secretase. In xPharm: The Comprehensive Pharmacology Reference; Enna, S.J., Bylund, D.B., Eds.; Elsevier: Amsterdam, The Netherlands, 2007; pp. 1–10. [Google Scholar]
  149. Reddy, P.H.; Williams, J.; Smith, F.; Bhatti, J.S.; Kumar, S.; Vijayan, M.; Kandimalla, R.; Kuruva, C.S.; Wang, R.; Manczak, M.; et al. MicroRNAs, Aging, Cellular Senescence, and Alzheimer’s Disease. Prog. Mol. Biol. Transl. Sci. 2017, 146, 127–171. [Google Scholar] [CrossRef]
  150. Haass, C.; Kaether, C.; Thinakaran, G.; Sisodia, S. Trafficking and proteolytic processing of APP. Cold Spring Harb. Perspect. Med. 2012, 2, a006270. [Google Scholar] [CrossRef]
  151. Talwar, P.; Gupta, R.; Kushwaha, S.; Agarwal, R.; Saso, L.; Kukreti, S.; Kukreti, R. Viral Induced Oxidative and Inflammatory Response in Alzheimer’s Disease Pathogenesis with Identification of Potential Drug Candidates: A Systematic Review using Systems Biology Approach. Curr. Neuropharmacol. 2019, 17, 352–365. [Google Scholar] [CrossRef]
  152. Medeiros, R.; Baglietto-Vargas, D.; LaFerla, F.M. The role of tau in Alzheimer’s disease and related disorders. CNS Neurosci. Ther. 2011, 17, 514–524. [Google Scholar] [CrossRef]
  153. Burley, S.K.; Berman, H.M.; Kleywegt, G.J.; Markley, J.L.; Nakamura, H.; Velankar, S. Protein Data Bank (PDB): The Single Global Macromolecular Structure Archive. Methods Mol. Biol. 2017, 1607, 627–641. [Google Scholar] [CrossRef] [Green Version]
  154. Guerreiro, R.J.; Gustafson, D.R.; Hardy, J. The genetic architecture of Alzheimer’s disease: Beyond APP, PSENs and APOE. Neurobiol. Aging 2012, 33, 437–456. [Google Scholar] [CrossRef]
  155. Cuccaro, M.L.; Carney, R.M.; Zhang, Y.; Bohm, C.; Kunkle, B.W.; Vardarajan, B.N.; Whitehead, P.L.; Cukier, H.N.; Mayeux, R.; George-Hyslop, P.S. SORL1 mutations in early-and late-onset Alzheimer disease. Neurol. Genet. 2016, 2, e116. [Google Scholar] [CrossRef]
  156. D’Argenio, V.; Sarnataro, D. New insights into the molecular bases of familial Alzheimer’s disease. J. Pers. Med. 2020, 10, 26. [Google Scholar] [CrossRef]
  157. Higgins, L.S.; Catalano, R.; Quon, D.; Cordell, B. Transgenic mice expressing human beta-APP751, but not mice expressing beta-APP695, display early Alzheimer’s disease-like histopathology. Ann. N. Y. Acad. Sci. 1993, 695, 224–227. [Google Scholar] [CrossRef]
  158. Hooli, R.E.T.B. The Genetic Basis of Alzheimer’s Disease: Findings from Genome-Wide Studies. In State, Genomics, Circuits, and Pathways in Clinical Neuropsychiatry; Lehner, B.L.M.T., Matthew, W., Eds.; Academic Press: Cambridge, MA, USA, 2016; pp. 547–571. [Google Scholar]
  159. Eratne, D.; Loi, S.M.; Farrand, S.; Kelso, W.; Velakoulis, D.; Looi, J.C. Alzheimer’s disease: Clinical update on epidemiology, pathophysiology and diagnosis. Australas. Psychiatry 2018, 26, 347–357. [Google Scholar] [CrossRef]
  160. Sevigny, J.; Chiao, P.; Bussiere, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
  161. Kastanenka, K.V.; Bussiere, T.; Shakerdge, N.; Qian, F.; Weinreb, P.H.; Rhodes, K.; Bacskai, B.J. Immunotherapy with Aducanumab Restores Calcium Homeostasis in Tg2576 Mice. J. Neurosci. 2016, 36, 12549–12558. [Google Scholar] [CrossRef]
  162. Padda, I.S.; Parmar, M. Aducanumab; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  163. Arndt, J.W.; Qian, F.; Smith, B.A.; Quan, C.; Kilambi, K.P.; Bush, M.W.; Walz, T.; Pepinsky, R.B.; Bussiere, T.; Hamann, S.; et al. Structural and kinetic basis for the selectivity of aducanumab for aggregated forms of amyloid-beta. Sci. Rep. 2018, 8, 6412. [Google Scholar] [CrossRef]
  164. Cummings, J.; Aisen, P.; Lemere, C.; Atri, A.; Sabbagh, M.; Salloway, S. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering. Alzheimers Res. Ther. 2021, 13, 98. [Google Scholar] [CrossRef]
  165. Fujimoto, T.; Matsushita, Y.; Gouda, H.; Yamaotsu, N.; Hirono, S. In silico multi-filter screening approaches for developing novel β-secretase inhibitors. Bioorg. Med. Chem. Lett. 2008, 18, 2771–2775. [Google Scholar] [CrossRef]
  166. AID 332040. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/332040 (accessed on 10 April 2022).
  167. Huang, D.; Luthi, U.; Kolb, P.; Edler, K.; Cecchini, M.; Audetat, S.; Barberis, A.; Caflisch, A. Discovery of cell-permeable non-peptide inhibitors of beta-secretase by high-throughput docking and continuum electrostatics calculations. J. Med. Chem. 2005, 48, 5108–5111. [Google Scholar] [CrossRef]
  168. AID 239369. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/239369 (accessed on 10 April 2022).
  169. AID 240366. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/240366 (accessed on 10 April 2022).
  170. AID 240368. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/240368 (accessed on 10 April 2022).
  171. Rajamani, R.; Reynolds, C.H. Modeling the binding affinities of beta-secretase inhibitors: Application to subsite specificity. Bioorg. Med. Chem. Lett. 2004, 14, 4843–4846. [Google Scholar] [CrossRef]
  172. AID 238396. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/238396 (accessed on 10 April 2022).
  173. AID 240791. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/240791 (accessed on 10 April 2022).
  174. Jeon, S.Y.; Bae, K.; Seong, Y.H.; Song, K.S. Green tea catechins as a BACE1 (beta-secretase) inhibitor. Bioorg. Med. Chem. Lett. 2003, 13, 3905–3908. [Google Scholar] [CrossRef]
  175. AID 44249. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/44249 (accessed on 10 April 2022).
  176. AID 44250. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/44250 (accessed on 10 April 2022).
  177. AID 242654. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/242654 (accessed on 10 April 2022).
  178. Hom, R.K.; Fang, L.Y.; Mamo, S.; Tung, J.S.; Guinn, A.C.; Walker, D.E.; Davis, D.L.; Gailunas, A.F.; Thorsett, E.D.; Sinha, S.; et al. Design and synthesis of statine-based cell-permeable peptidomimetic inhibitors of human beta-secretase. J. Med. Chem. 2003, 46, 1799–1802. [Google Scholar] [CrossRef]
  179. AID 44238. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/44238 (accessed on 10 April 2022).
  180. Mekala, S.; Nelson, G.; Li, Y.M. Recent developments of small molecule gamma-secretase modulators for Alzheimer’s disease. RSC Med. Chem 2020, 11, 1003–1022. [Google Scholar] [CrossRef]
  181. AID 1678948. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678948 (accessed on 10 April 2022).
  182. AID 1678949. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678949 (accessed on 10 April 2022).
  183. AID 1678953. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678953 (accessed on 10 April 2022).
  184. AID 1678954. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678954 (accessed on 10 April 2022).
  185. AID 1678955. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678955 (accessed on 10 April 2022).
  186. AID 1678967. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678967 (accessed on 10 April 2022).
  187. AID 1678968. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678968 (accessed on 10 April 2022).
  188. AID 1678971. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/1678971 (accessed on 10 April 2022).
  189. Czirr, E.; Leuchtenberger, S.; Dorner-Ciossek, C.; Schneider, A.; Jucker, M.; Koo, E.H.; Pietrzik, C.U.; Baumann, K.; Weggen, S. Insensitivity to Aβ42-lowering Nonsteroidal Anti-inflammatory Drugs and γ-Secretase Inhibitors Is Common among Aggressive Presenilin-1 Mutations. J. Biol. Chem. 2007, 282, 24504–24513. [Google Scholar] [CrossRef]
  190. AID 359781. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/359781 (accessed on 10 April 2022).
  191. Bakshi, P.; Jin, C.; Broutin, P.; Berhane, B.; Reed, J.; Mullan, M. Structural optimization of a CXCR2-directed antagonist that indirectly inhibits γ-secretase and reduces Aβ. Bioorg. Med. Chem. 2009, 17, 8102–8112. [Google Scholar] [CrossRef]
  192. AID 452857. PubChem Bioassay Record. Available online: https://pubchem.ncbi.nlm.nih.gov/bioassay/452857 (accessed on 10 April 2022).
  193. Liu, X.Y.; Zhang, N.; Zhang, S.X.; Xu, P. Potential new therapeutic target for Alzheimer’s disease: Glucagon-like peptide-1. Eur. J. Neurosci. 2021, 54, 7749–7769. [Google Scholar] [CrossRef]
  194. Jia, Q.; Deng, Y.; Qing, H. Potential therapeutic strategies for Alzheimer’s disease targeting or beyond beta-amyloid: Insights from clinical trials. BioMed. Res. Int. 2014, 2014, 837157. [Google Scholar] [CrossRef]
  195. Yu, T.W.; Lane, H.Y.; Lin, C.H. Novel Therapeutic Approaches for Alzheimer’s Disease: An Updated Review. Int. J. Mol. Sci. 2021, 22, 8208. [Google Scholar] [CrossRef]
  196. Guo, L.; Xu, J.; Du, Y.; Wu, W.; Nie, W.; Zhang, D.; Luo, Y.; Lu, H.; Lei, M.; Xiao, S.; et al. Effects of gut microbiota and probiotics on Alzheimer’s disease. Transl. Neurosci. 2021, 12, 573–580. [Google Scholar] [CrossRef]
  197. Kruger, J.F.; Hillesheim, E.; Pereira, A.; Camargo, C.Q.; Rabito, E.I. Probiotics for dementia: A systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 2021, 79, 160–170. [Google Scholar] [CrossRef] [PubMed]
  198. Walsham, N.E.; Sherwood, R.A. Fecal calprotectin in inflammatory bowel disease. Clin. Exp. Gastroenterol. 2016, 9, 21–29. [Google Scholar] [CrossRef]
  199. Wang, C.; Klechikov, A.G.; Gharibyan, A.L.; Warmlander, S.K.; Jarvet, J.; Zhao, L.; Jia, X.; Narayana, V.K.; Shankar, S.K.; Olofsson, A.; et al. The role of pro-inflammatory S100A9 in Alzheimer’s disease amyloid-neuroinflammatory cascade. Acta Neuropathol. 2014, 127, 507–522. [Google Scholar] [CrossRef]
  200. Zhang, C.; Liu, Y.; Gilthorpe, J.; van der Maarel, J.R. MRP14 (S100A9) protein interacts with Alzheimer beta-amyloid peptide and induces its fibrillization. PLoS ONE 2012, 7, e32953. [Google Scholar] [CrossRef]
  201. Leblhuber, F.; Geisler, S.; Steiner, K.; Fuchs, D.; Schutz, B. Elevated fecal calprotectin in patients with Alzheimer’s dementia indicates leaky gut. J. Neural Transm. 2015, 122, 1319–1322. [Google Scholar] [CrossRef] [PubMed]
  202. Marizzoni, M.; Provasi, S.; Cattaneo, A.; Frisoni, G.B. Microbiota and neurodegenerative diseases. Curr. Opin. Neurol. 2017, 30, 630–638. [Google Scholar] [CrossRef]
  203. Potgieter, M.; Bester, J.; Kell, D.B.; Pretorius, E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol. Rev. 2015, 39, 567–591. [Google Scholar] [CrossRef]
  204. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
  205. Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability--a new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef] [Green Version]
  206. Konig, J.; Wells, J.; Cani, P.D.; Garcia-Rodenas, C.L.; MacDonald, T.; Mercenier, A.; Whyte, J.; Troost, F.; Brummer, R.J. Human Intestinal Barrier Function in Health and Disease. Clin. Transl. Gastroenterol. 2016, 7, e196. [Google Scholar] [CrossRef]
  207. Choi, V.M.; Herrou, J.; Hecht, A.L.; Teoh, W.P.; Turner, J.R.; Crosson, S.; Wardenburg, J.B. Activation of Bacteroides fragilis toxin by a novel bacterial protease contributes to anaerobic sepsis in mice. Nat. Med. 2016, 22, 563–567. [Google Scholar] [CrossRef]
  208. Yu, Y.; Zhao, F. Microbiota-gut-brain axis in autism spectrum disorder. J. Genet. Genom. 2021, 48, 755–762. [Google Scholar] [CrossRef]
  209. Lanoiselee, H.M.; Nicolas, G.; Wallon, D.; Rovelet-Lecrux, A.; Lacour, M.; Rousseau, S.; Richard, A.C.; Pasquier, F.; Rollin-Sillaire, A.; Martinaud, O.; et al. APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: A genetic screening study of familial and sporadic cases. PLoS Med. 2017, 14, e1002270. [Google Scholar] [CrossRef]
  210. Chen, C.J.; Ou, Y.C.; Li, J.R.; Chang, C.Y.; Pan, H.C.; Lai, C.Y.; Liao, S.L.; Raung, S.L.; Chang, C.J. Infection of pericytes in vitro by Japanese encephalitis virus disrupts the integrity of the endothelial barrier. J. Virol. 2014, 88, 1150–1161. [Google Scholar] [CrossRef]
  211. Kim, K.S. How pathogens penetrate the blood-brain barrier: To reach the central nervous system, meningitis-causing strains of E. coli and other pathogens exploit microbial and host factors. Microbe 2014, 9, 487–492. [Google Scholar] [CrossRef]
  212. Xi, J.; Ding, D.; Zhu, H.; Wang, R.; Su, F.; Wu, W.; Xiao, Z.; Liang, X.; Zhao, Q.; Hong, Z.; et al. Disturbed microbial ecology in Alzheimer’s disease: Evidence from the gut microbiota and fecal metabolome. BMC Microbiol. 2021, 21, 226. [Google Scholar] [CrossRef] [PubMed]
  213. Bhaskaran, N.; Quigley, C.; Paw, C.; Butala, S.; Schneider, E.; Pandiyan, P. Role of Short Chain Fatty Acids in Controlling Tregs and Immunopathology During Mucosal Infection. Front. Microbiol. 2018, 9, 1995. [Google Scholar] [CrossRef]
  214. Zhang, Y.; Geng, R.; Tu, Q. Gut microbial involvement in Alzheimer’s disease pathogenesis. Aging 2021, 13, 13359–13371. [Google Scholar] [CrossRef]
  215. Dhillon, S. Aducanumab: First Approval. Drugs 2021, 81, 1437–1443. [Google Scholar] [CrossRef]
  216. Vaz, M.; Silva, V.; Monteiro, C.; Silvestre, S. Role of Aducanumab in the Treatment of Alzheimer’s Disease: Challenges and Opportunities. Clin. Interv. Aging 2022, 17, 797–810. [Google Scholar] [CrossRef]
  217. Fleisher, A.S.; Raman, R.; Siemers, E.R.; Becerra, L.; Clark, C.M.; Dean, R.A.; Farlow, M.R.; Galvin, J.E.; Peskind, E.R.; Quinn, J.F.; et al. Phase 2 safety trial targeting amyloid beta production with a gamma-secretase inhibitor in Alzheimer disease. Arch. Neurol. 2008, 65, 1031–1038. [Google Scholar] [CrossRef] [PubMed]
  218. Salloway, S.; Sperling, R.; Fox, N.C.; Blennow, K.; Klunk, W.; Raskind, M.; Sabbagh, M.; Honig, L.S.; Porsteinsson, A.P.; Ferris, S.; et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 322–333. [Google Scholar] [CrossRef]
  219. Wischik, C.; Staff, R. Challenges in the conduct of disease-modifying trials in AD: Practical experience from a phase 2 trial of Tau-aggregation inhibitor therapy. J. Nutr. Health Aging 2009, 13, 367–369. [Google Scholar] [CrossRef]
  220. Doody, R.S.; Farlow, M.; Aisen, P.S. Phase 3 trials of solanezumab and bapineuzumab for Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 1460. [Google Scholar] [CrossRef]
  221. Khan, T.K.; Alkon, D.L. Alzheimer’s Disease Cerebrospinal Fluid and Neuroimaging Biomarkers: Diagnostic Accuracy and Relationship to Drug Efficacy. J. Alzheimers Dis. 2015, 46, 817–836. [Google Scholar] [CrossRef]
  222. Saumier, D.; Aisen, P.S.; Gauthier, S.; Vellas, B.; Ferris, S.H.; Duong, A.; Suhy, J.; Oh, J.; Lau, W.; Garceau, D.; et al. Lessons learned in the use of volumetric MRI in therapeutic trials in Alzheimer’s disease: The ALZHEMED (Tramiprosate) experience. J. Nutr. Health Aging 2009, 13, 370–372. [Google Scholar] [CrossRef]
  223. Braak, H.; Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991, 82, 239–259. [Google Scholar] [CrossRef]
  224. Mirra, S.S.; Heyman, A.; McKeel, D.; Sumi, S.M.; Crain, B.J.; Brownlee, L.M.; Vogel, F.S.; Hughes, J.P.; van Belle, G.; Berg, L. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology 1991, 41, 479–486. [Google Scholar] [CrossRef]
  225. Guo, T.; Zhang, D.; Zeng, Y.; Huang, T.Y.; Xu, H.; Zhao, Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol. Neurodegener. 2020, 15, 1–37. [Google Scholar] [CrossRef]
  226. Vigasova, D.; Nemergut, M.; Liskova, B.; Damborsky, J. Multi-pathogen infections and Alzheimer’s disease. Microb. Cell Fact. 2021, 20, 25. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Graphical presentation depicting certain differences in gut and brain composition between healthy and unhealthy individuals. Infection with microbes inside the gut is responsible for the microbial transfer into the blood stream, and this may lead to neurodegeneration, neuroinflammation, neurodysfunction, and dysbiosis. After passing through the blood–brain barrier, a microorganism may affect APP processing, and this causes an increase in amyloid beta (Aβ) protein deposition between neurons. Inflammation among neurons and excessive phosphorylation promote tau protein disintegration, which culminates in neuronal damage and the onset of Alzheimer’s disease (AD). Dysbiosis is caused by microorganism infection, resulting in the elimination of helpful bacteria such as Firmicutes.
Figure 1. Graphical presentation depicting certain differences in gut and brain composition between healthy and unhealthy individuals. Infection with microbes inside the gut is responsible for the microbial transfer into the blood stream, and this may lead to neurodegeneration, neuroinflammation, neurodysfunction, and dysbiosis. After passing through the blood–brain barrier, a microorganism may affect APP processing, and this causes an increase in amyloid beta (Aβ) protein deposition between neurons. Inflammation among neurons and excessive phosphorylation promote tau protein disintegration, which culminates in neuronal damage and the onset of Alzheimer’s disease (AD). Dysbiosis is caused by microorganism infection, resulting in the elimination of helpful bacteria such as Firmicutes.
Cimb 44 00314 g001
Figure 2. The mechanisms behind the neurological consequences of microorganisms affecting the brain. The gut microbiome imbalance affects the shortage of microbiome diversity and leads to an enhancement of intestinal permeability. Released neurotoxic agents such as proinflammatory cytokines from microorganisms can transmit to the brain through interactions between the intestine’s enteric nervous system and central nervous system. This results in the breakdown of tight junctions of the blood–brain barrier and symptoms such as Aβ aggregation (amyloid plaques and oxidative stress), neuro-inflammation, tau hyperphosphorylation, and memory loss. Lastly, it can contribute to the pathogenesis of neurological diseases like AD.
Figure 2. The mechanisms behind the neurological consequences of microorganisms affecting the brain. The gut microbiome imbalance affects the shortage of microbiome diversity and leads to an enhancement of intestinal permeability. Released neurotoxic agents such as proinflammatory cytokines from microorganisms can transmit to the brain through interactions between the intestine’s enteric nervous system and central nervous system. This results in the breakdown of tight junctions of the blood–brain barrier and symptoms such as Aβ aggregation (amyloid plaques and oxidative stress), neuro-inflammation, tau hyperphosphorylation, and memory loss. Lastly, it can contribute to the pathogenesis of neurological diseases like AD.
Cimb 44 00314 g002
Figure 3. Graph depicting the methodology employed to detect the microorganism involved in causing Alzheimer’s disease. According to the data, most of the microorganisms were detected using the PCR-based analysis method, whereas very few were detected using RNA sequencing technology.
Figure 3. Graph depicting the methodology employed to detect the microorganism involved in causing Alzheimer’s disease. According to the data, most of the microorganisms were detected using the PCR-based analysis method, whereas very few were detected using RNA sequencing technology.
Cimb 44 00314 g003
Figure 4. Permeation of microbial byproducts (toxins) and live microorganisms through the mucosal layer as part of the disruption in the BBB; result in disassembly, accumulation, and inflammation between neurons, which lead to AD.
Figure 4. Permeation of microbial byproducts (toxins) and live microorganisms through the mucosal layer as part of the disruption in the BBB; result in disassembly, accumulation, and inflammation between neurons, which lead to AD.
Cimb 44 00314 g004
Table 1. Microorganisms and the research methodology used in determining a role in Alzheimer’s disease (AD).
Table 1. Microorganisms and the research methodology used in determining a role in Alzheimer’s disease (AD).
S.No.Name of
Microorganisms
Role in ADMethodologyYearReference
1Helicobacter pylori▪ Upregulation of Tau protein
▪ Neuroinflammation
▪ Neurotoxicity
▪ Immunochemical analysis
▪ Statistical analysis
▪ ELISA
▪ PCR based analysis
2005, 2013,
2015
[92,102,103,104]
2ActinobacteriaModulation of specific gene expressions16S rRNA sequencing2017[105]
3FirmicutesCerebral Aβ amyloidosis16S rRNA sequencing2017[105,106]
4ProteobacteriaRelease of proinflammatory cytokines16S rRNA sequencing2017[105]
5TreponemadenticolaInflammation in brain▪ 16S rDNA sequencing
▪ ELISA
2009, 2012, 2019, 2020[107,108,109,110]
6Gingivitis bacteria▪ Alteration of tau and ubiquitin
▪ Leading to chronic inflammation
16S rDNA sequencing2019[109]
7Porphyromonas
gingivalis
▪ Cleavage of Tau protein
▪ Phosphorylate neuronal tau
Serological studies (ELISA)2009[107,111]
8Tannerella forsythiaInflammation▪ PCR-based analysis
▪ Immunochemical analysis
2019[31]
9Toxoplasma gondii▪ Inflammation in CNS
▪ Activation of T cells
▪ Oxidative stress
Immunochemical analysis2015[112]
10Herpes simplex Virus
(HSV)
▪ Neurotrophic
▪ Neuroinvasive
▪ Brain infection
▪ Encephalitis in adults
▪ Meningitis in neonates
▪ Lesions in infected individuals’ brains
▪ ELISA
▪ PCR-based analysis
▪ RNA sequencing
▪ Statistical analysis
2015, 2003,
2002, 2014,
2018
[56,101,104,113,114,115]
11Saccharomyces
cerevisiae
Oxidative stress▪ Immunofluorescence analysis
▪ Slot-blot analysis,
▪ Proteomic analysis
▪ PCR- based analysis
2014, 2017[58,59,116,117]
12Malassezia speciesNeuroinflammatory responsethrough T-cell activation▪ PCR-based analysis
▪ Immunochemical analysis
▪ Immunofluorescence analysis
▪ Slot- blot analysis
2014[58,59]
13Candida speciesActivation of NF-κB leading to proinflammatory cytokines IL-6, IL-1,
and TNF
▪ Proteomic analysis
▪ PCR-based analysis
▪ Immunochemical analysis
▪ Slot- blot analysis
▪ NGS
2014, 2017[58,59,116,117]
14Cladosporium
cryptococcus
Neuroinflammation▪ PCR-based analysis
▪ Slot- blot analysis
▪ NGS
2014, 2017[58,59,116,117]
Table 2. Mouse and Drosophila models used to identify a role for various treatments in AD.
Table 2. Mouse and Drosophila models used to identify a role for various treatments in AD.
S.No.Name of
Microorganisms
Treatment to ModelResult of ExperimentYearReference
1APOE−/− micePorphyromonas gingivalis active invasionActivation in APOE−/− mice brain2015[120]
2APP/PS1 miceAntibiotic treatment to Tg mice▪ Changes in composition of intestinal microbiome
▪ Decrease concentration in Aβ deposition and soluble Aβ increased
▪ Reactive gliosis decreased
▪ Expansion of Lachnospiraceae
▪ Aβ deposition in aged Tg mice is reduced
▪ Plaque-localized microglia and astrocytes reduced in antibiotic-exposed mice
2016, 2017,
2018,
[121,122,123,124]
35XFAD miceComposition of fecal microbiota changed along with age▪ Enzyme trypsin in human fecal matter decreased
▪ APP expression seen in the gut tissues.
2017[118]
43XTg-AD miceMice treated with probiotics ▪ Concentration of plasma influenced in the case of inflammatory cytokines and gut hormones
▪ Accumulation of Aβ aggregates and brain damage reduced
2017[119]
5AD mouse model
(ICV injection of Aβ)
Bifidobacterium breve strain A1given orallyFunction of hippocampus enhanced2017[125]
6AD rat model
(IP injection of D- galactose)
Mice treated with Lactobacillus plantarum MTCC 1325Acetylcholine level is restored, formation of Aβ plaque attenuated2017[126]
7AD rat model
(intrahippocampal injection of Aβ)
Mice treated with Lactobacillus and BifidobacteriumLearning problems and oxidative stress observed2018[127]
8Transgenic flies: DrosophilaDrosophila exposed to Enterobacteria infection▪ Neuroinflammation is reduced when hemocytes are genetically depleted
▪ Neurodegeneration reduced
2017[128]
95XFAD miceMulti-antibiotic treatment
- Gentamicin (0.1251 mg/mL)
- Vancomycin (0.0635 mg/mL)
- Metronidazole (0.25 mg/mL)
- Neomycin (0.0635 mg/mL)
- Ampicillin (0.1251 mg/mL)
- Kanamycin (0.3753 mg/mL)
- Colistin (7,506,000 U/mL)
- Cefoperazone (0.1251 mg/mL)
▪ Manipulation of the gut microbiome
▪ Mice revealed antidiabetic properties
▪ Effect on nest building quality
2021[129]
Table 3. IC50, EC50, and Ki values of bioactive beta-secretase inhibitors, a therapeutic target for AD (from various studies of the last 5 years).
Table 3. IC50, EC50, and Ki values of bioactive beta-secretase inhibitors, a therapeutic target for AD (from various studies of the last 5 years).
No.IUPAC Names of CompoundsCIDSIDAIDIC50
(µM)
EC50
(µM)
Ki Value (µM)Reference
11-N-[(2S,3S,5R)-3-amino-6-(4-fluoroani-li-no)-5-methyl-6-oxo-1-phenylhexan-2-yl] -3-N,3-N-dipropyl- benzene-1,3-dicarboxamide5494423103477859332040 0.026[165,166]
23-benzoyl-N-[(2S,3R)-4-(cyclopropylam-ino)-3-hydroxy-1-phenylbutan-2-yl]-5-[methyl(methylsulfonyl)amino]benzamide53270631034784103320400.098 [165,166]
33-N-[(2S,3R)-4-(cyclopropylamino)-3-hydroxy-1-phenylbutan-2-yl]-5-[me-thyl(methylsulfonyl)amino]-1-N-[(1R)-1-phenylethyl]benzene-1,3-dicarboxamide52875321034928193320400.015 [165,166]
4[3-[2-(5-aminopentylamino)-2-oxoethoxy]-5-[[(1R)-1-(4-fluorophenyl)ethyl]carbamoyl]phenyl] phenylmethanesulfonate 4487721035119353320401.4 [165,166]
51-carbazol-9-yl-3-[4-(3-carbazol-9-yl-2-hydroxypropyl)piperazin-1-yl]propan-2-ol27290221035641703320407 [165,166]
61-[4-(3-carbazol-9-yl-2-hydroxypropyl)piperazin-1-yl]-3-(3,6-dichlorocarbazol-9-yl)propan-2-ol28023721035642153320405 [165,166]
7 1-(3-acetylphenyl)-3-(3-ethylsulfanyl-1,2,4-thiadiazol-5-yl)urea 11198061103459736239369 28.8[167,168]
85-[[3,5-bis(trifluoromethyl)phenyl]car-bam-oylamino]-2-(dimethylamino)-N-(3-morpholin-4-ylpropyl)benzamide4243074103459765239369 8.1[167,168]
95-[[3,5-bis(trifluoromethyl)phenyl]car-bam-oylamino]-2-(dimethylamino)-N-(3-methoxypropyl)benzamide5040262103459791239369 9.4[167,168]
105-[(3-chlorophenyl)carbamoylamino]-2-(dimethylamino)-N-(3-ethoxypropyl)benzamide3948694103460050239369 9.8[167,168]
111-(3-acetylphenyl)-3-(3-ethylsulfanyl-1,2,4-thiadiazol-5-yl)urea11198061103459736240366 0.0114 [167,169]
125-[[3,5-bis(trifluoromethyl)phenyl]car-bam-oylamino]-2-(dimethylamino)-N-(3-methoxypropyl)benzamide5040262103459791240366 27 [167,169]
135-[(3-chlorophenyl)carbamoylamino]-2-(dimethylamino)-N-(3-ethoxypropyl)benzamide3948694103460050240366 0.0233 [167,169]
145-[[3,5-bis(trifluoromethyl)phenyl]car-bam-oylamino]-2-(dimethylamino)-N-(3-morpholin-4-ylpropyl)benzamide4243074103459765240368 0.0035 [167,170]
15tert-butyl N-[1-[[(4S)-8-[[1-(benzyl-amino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-4-methylsulfanyl-1-oxobutan-2-yl]carbamate44305543103256754238396 5.808[171,172]
16tert-butyl N-[(2S)-1-[[1-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-4-methylsulfonyl-1-oxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate44305867103257369238396 0.008[171,172]
17tert-butyl N-[(2S)-1-[[1-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-4-methylsulfanyl-1-oxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate44305868103257370238396 0.0025[171,172]
18tert-butyl N-[(2S)-1-[[1-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-3-methylsulfonyl-1-oxopropan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate44305869103257371238396 0.0094[171,172]
19tert-butyl N-[(2S)-1-[[4-amino-1-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1,4-dioxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate 44305870103257372238396 0.0059[171,172]
20tert-butyl N-[(2S)-1-[[4-amino-1-[[(4S)-8-[[1-(benzylamino)-1-oxopropan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1,4-dioxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate44305918103257473238396 0.0614[171,172]
21tert-butyl N-[(2S)-1-[[1-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-3-methylsulfanyl-1-oxopropan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]carbamate44305946103257506238396 0.0501[171,172]
22(4S)-4-amino-5-[[(2S)-1-[[(2S)-4-amino-1-[[(4S,5S,7R)-8-[[(2S)-1-[[(2S)-4-carboxy-1-[[(1S)-1-carboxy-2-phenylethyl]amino]-1-oxobutan-2-yl]amino]-1-oxopropan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1,4-dioxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-5-oxopentanoic acid445649103282992238396 0.0016[171,172]
23tert-butyl N-[(3S)-1-amino-5-[[(4S)-8-[[1-(benzylamino)-1-oxopropan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1,4-dioxopentan-3-yl]carbamate44394717103451640238396 22.423[171,172]
24tert-butyl N-[(3S)-1-amino-5-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1,4-dioxopentan-3-yl]carbamate 44394718103451641238396 3.134[171,172]
25tert-butyl N-[(2R)-4-[[(4S)-8-[[1-(benzylamino)-3-methyl-1-oxobutan-2-yl]amino]-5-hydroxy-2,7-dimethyl-8-oxooctan-4-yl]amino]-1-methylsulfonyl-3-oxobutan-2-yl]carbamate44394719103451642238396 1.129[171,172]
26 (2R,5S)-5-[[(2S)-2-[[(2R,4S,5S)-5-[[(2S)-2-[[(2S)-2-[[(2S)-2-amino-4-carboxybutanoyl]amino]-4-methylpentanoyl]amino]-3-carboxypropanoyl]amino]-4-hydroxy-2,7-dimethyloctanoyl]amino]-3-methylbutanoyl]amino]-2-benzyl-4-oxooctanedioic acid 44394812103451744238396 0.00032[171,172]
271-N-[(2S,3S,5R)-5-(benzylcarbamoyl)-1-(3,5-difluorophenyl)-3-hydroxyheptan-2-yl]-3-N,3-N-dipropylbenzene-1,3-dicarboxamide101216281032229032407911.4 [171,173]
281-N-[(2S,3S,5R)-1-(3,5-difluorophenyl)-5-[[(3S,5R)-3,5-dimethoxycyclohexyl]carbamoyl]-3-hydroxyheptan-2-yl]-3-N,3-N-dipropylbenzene-1,3-dicarboxamide113543221032229042407916.6 [171,173]
295-[[(2R,4S,5S)-6-(3,5-difluorophenyl)-5-[[3-(dipropylcarbamoyl)benzoyl]amino]-2-ethyl-4-hydroxyhexanoyl]amino]pentanoic acid102825551032229352407910.4 [171,173]
303-[[(2R,4S,5S)-6-(3,5-difluorophenyl)-5-[[3-(dipropylcarbamoyl)benzoyl]amino]-2-ethyl-4-hydroxyhexanoyl]amino]propanoic acid 10167359 2407912.8- [171,173]
314-[[[(2R,4S,5S)-6-(3,5-difluorophenyl)-5-[[3-(dipropylcarbamoyl)benzoyl]amino]-2-ethyl-4-hydroxyhexanoyl]amino]methyl]cyclohexane-1-carboxylic acid98098111032231592407910.05 [171,173]
32[(2S,3S)-2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate65056103359341442494.5 [95,96]
33(2R,3S)-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol65084103359373442492.5 [174,175]
34[(2R,3S)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate5276890103359448442491.8 [174,175]
35(2S,3S)-2-(3,4-dihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol1822321033594664424928 [174,175]
36 (2S,3S)-2-(3,4-dihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol 1822321033594664424923 [174,175]
37 (2S,3S)-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol10425234103359508442492.4 [174,175]
38[(2S,3R)-2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate6419835103359509442496 [174,175]
39[(2S,3S)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate2824823103359580442491.6 [174,175]
40(2S,3R)-2-(3,4-dihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol731601034747754424930 [174,175]
41(2R,3S)-2-(3,4-dihydroxyphenyl)-3,4-dihydro-2H-chromene-3,5,7-triol90641230947114424935 [174,175]
42[(2S,3S)-2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate 6505610335934144250 5.3[174,176]
43[(2R,3S)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate 527689010335944844250 0.17[174,176]
44[(2S,3S)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-chromen-3-yl] 3,4,5-trihydroxybenzoate282482310335958044250 0.21[174,176]
455-[[3,5-bis(trifluoromethyl)phenyl]carbamoylamino]-2-(dimethyl- amino)-N-(3-morpholin-4-ylpropyl)benzamide42430741034597652426540.0578 [167,177]
465-[(3-chlorophenyl)carbamoylamino]-2-(dimethylamino)-N-(3-ethoxypropyl)benzamide39486941034600502426540.097 [167,177]
47(4R)-4-[[(2R)-2-[[(2R)-2-[[(3S)-4-[(4-butoxybenzoyl)amino]-3-hydroxy-6-methylheptanoyl]amino]-3-methylbutanoyl]amino]propanoyl]amino]-5-[[(1R)-1-carboxy-2-phenylethyl]amino]-5-oxopentanoic acid 442965881032369754423827 [178,179]
48methyl 4-[[(2R)-2-[[(3S)-3-hydroxy-6-methyl-4-[[(2S)-2-[[(2S)-3-methyl-2-[(2-methylpropan-2-yl)oxycarbonylamino]butanoyl]amino]-4-methylsulfanylbutanoyl]amino]heptanoyl]amino]-3-methylbutanoyl]amino]butanoate 442964831032367544423847 [178,179]
493-[[(2R)-2-[[(3S)-3-hydroxy-6-methyl-4-[[(2S)-2-[[(2S)-3-methyl-2-[(2-methylpropan-2-yl)oxycarbonylamino]butanoyl]amino]-4-methylsulfanylbutanoyl]amino]heptanoyl]amino]-3-methylbutanoyl]amino]propanoic acid442964541032366964423830 [178,179]
504-[[[(2R)-2-[[(3S)-3-hydroxy-6-methyl-4-[[(2S)-2-[[(2S)-3-methyl-2-[(2-methylpropan-2-yl)oxycarbonylamino]butanoyl]amino]-4-methylsulfanylbutanoyl]amino]heptanoyl]amino]-3-methylbutanoyl]amino]methyl]benzoic acid44296453103236695442384 [178,179]
Table 4. IC50, EC50, and Ki values of bioactive gamma secretase inhibitors, a therapeutic target for AD (from various studies of the last 5 years).
Table 4. IC50, EC50, and Ki values of bioactive gamma secretase inhibitors, a therapeutic target for AD (from various studies of the last 5 years).
No.IUPAC Names of CompoundsCIDSIDAIDIC50
(µM)
EC50
(µM)
PMIDReference
1(2R)-2-[4-[(R)-(4-fluorophenyl)-(4-methylpiperidin-1-yl)methyl]-3-[4-(trifluoromethyl)phenyl]phenyl]propanoic acid534930011633265811678948 0.1533479693[180,181]
2(2R)-2-[4-[(R)-(4-fluorophenyl)-(4-methylpiperidin-1-yl)methyl]-3-[4-(trifluoromethyl)phenyl]phenyl]propanoic acid5349300116332658116789490.17 33479693[180,182]
35-[4-[8-(3,4-dichlorophenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyridin-3-yl]-2-methoxyphenyl]-2-methyl-1,3-oxazole6660806231839038316789530.24 33479693[180,183]
46-chloro-3′-[3-methoxy-4-(2-methyl-1,3-oxazol-5-yl)phenyl]-1-(2,2,2-trifluoroethyl)spiro[5H-4,1-benzoxazepine-3,8′-6,7-dihydro-5H-[1,2,4]triazolo[4,3-a]pyridine]-2-one 5752119933690300216789530.03 33479693[180,183]
53′-[3-methoxy-4-(2-methyl-1,3-oxazol-5-yl)phenyl]-1-(2,2,2-trifluoroethyl)-6-(trifluoromethyl)spiro[5H-4,1-benzoxazepine-3,8′-6,7-dihydro-5H-[1,2,4]triazolo[4,3-a]pyridine]-2-one12704994733690343916789530.031 33479693[180,183]
62-[(8R)-3-[3-methoxy-4-(2-methyl-1,3-oxazol-5-yl)phenyl]-8-(3,4,5-trifluorophenoxy)-6,7-dihydro-5H-[1,2,4]triazolo[4,3-a]pyridin-8-yl]propan-2-ol5683719633690344016789530.026 33479693[180,183]
72-[8-(3-chloro-4-fluorophenoxy)-3-[3-methoxy-4-(2-methyl-1,3-oxazol-5-yl)phenyl]-6,7-dihydro-5H-[1,2,4]triazolo[4,3-a]pyridin-8-yl]propan-2-ol 6660395833690514916789530.038 33479693[180,183]
88-[(3,4-difluorophenyl)methyl]-3-[3-methoxy-4-(2-methyl-1,3-oxazol-5-yl)phenyl]-N-(2,2,2-trifluoroethyl)-6,7-dihydro-5H-[1,2,4]triazolo[4,3-a]pyridine-8-carboxamide6660423633690684716789530.075 33479693[180,183]
98-(4-fluoro-2-methylphenyl)-N-(1-methylindazol-5-yl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine 14260777040466965916789541.2 33479693[180,184]
108-(4-fluoro-2-methylphenyl)-N-(1-methyl-4,5,6,7-tetrahydroindazol-5-yl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine 14260790040467193716789540.38 33479693[180,184]
11N-[3,3-difluoro-1-(3-methyl-1,2,4-thiadiazol-5-yl)piperidin-4-yl]-8-(2,3,4-trifluorophenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine5839789446151842816789540.143 33479693[180,184]
12(3E)-1-[(1S)-1-(4-fluorophenyl)ethyl]-3-[[3-methoxy-4-(4-methylimidazol-1-yl)phenyl]methylidene]piperidin-2-one 1156078710424493216789550.093 33479693[180,185]
13(2R)-2-[4-[(R)-(4-fluorophenyl)-(4-methylpiperidin-1-yl)methyl]-3-[4-(trifluoromethyl)phenyl]phenyl]propanoic acid 534930011633265811678967 0.14633479693[180,186]
14(2R)-2-[4-[(R)-(4-fluorophenyl)-(4-methylpiperidin-1-yl)methyl]-3-[4-(trifluoromethyl)phenyl]phenyl]propanoic acid5349300116332658116789680.064 33479693[180,187]
15(2R)-2-[3-chloro-4-(2,2,2-trifluoroethoxy)-5-[4-(trifluoromethyl)phenyl]phenyl]-3-cyclobutylpropanoic acid 7162304931838644116789710.067 33479693[180,188]
16(2S)-2-[[(2S)-2-(3,5-difluorophenyl)-2-hydroxyacetyl]amino]-N-[(7S)-5-methyl-6-oxo-7H-benzo[d][1]benzazepin-7-yl]propanamide10435235103537689359781 0.00011917573346[189,190]
171-(2-fluorophenyl)-3-(4-methoxy-2-nitrophenyl)urea299222710371961745285710 19853461[191,192]
181-(3,4-dimethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea 4622683810371956045285715 19853461[191,192]
191-(2-hydroxy-4-nitrophenyl)-3-(2,3,4-trifluorophenyl)urea 462268431037195694528578 19853461[191,192]
201-(3,5-dimethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea 462268531037195844528572 19853461[191,192]
211-(2-ethyl-6-methylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea462268571037195934528577 19853461[191,192]
221-(2-ethyl-6-propan-2-ylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea462268581037195944528573 19853461[191,192]
231-(2-fluorophenyl)-3-(2-methoxy-4-nitrophenyl)urea3104951037196054528575 19853461[191,192]
241-(3-fluorophenyl)-3-(2-methoxy-4-nitrophenyl)urea458096410371960645285712 19853461[191,192]
251-(2-hydroxy-4-nitrophenyl)-3-(2,4,5-trifluorophenyl)urea4622686610371960745285710 19853461[191,192]
261-(2-hydroxy-4-nitrophenyl)-3-(2-methylphenyl)urea220113191037196084528572 19853461[191,192]
271-(2,5-dimethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea4622683710371955945285710 19853461[191,192]
281-(2-ethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea211847951037196184528570.09 19853461[191,192]
291-(2-hydroxy-4-nitrophenyl)-3-(2-propylphenyl)urea461964161037196194528570.1 19853461[191,192]
301-(2-hydroxy-4-nitrophenyl)-3-[2-(2-methylpropyl)phenyl]urea462268741037196204528571 19853461[191,192]
311-(3-ethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea462268841037196344528574 19853461[191,192]
321-(4-ethylphenyl)-3-(2-hydroxy-4-nitrophenyl)urea462268851037196354528573 19853461[190,191]
331-(2-hydroxy-4-nitrophenyl)-3-(2-methyl-6-propan-2-ylphenyl)urea462268861037196364528575 19853461[190,191]
341-(2,4-dibromophenyl)-3-(2-hydroxy-4-nitrophenyl)urea 220113361037196494528570.8 19853461[191,192]
35 1-(2,4-dichlorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea220113101037196504528570.3 19853461[191,192]
361-(2-hydroxy-4-nitrophenyl)-3-(2,3,4-trichlorophenyl)urea462268141037195274528573 19853461[191,192]
371-(2-bromophenyl)-3-(2-hydroxy-4-nitrophenyl)urea38546661035476444528570.5 19853461[191,192]
381-[2,6-di(propan-2-yl)phenyl]-3-(2-hydroxy-4-nitrophenyl)urea 462267721037194674528578 19853461[191,192]
391-(2,4-difluorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea4622678610371948345285715 19853461[191,192]
401-(2,5-dichlorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea45927491037194844528570.5 19853461[191,192]
41 1-(3,4-dichlorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea462268041037195084528570.7 19853461[191,192]
421-(3,4-difluorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea 462268051037195094528578 19853461[191,192]
43 1-(2-hydroxy-4-nitrophenyl)-3-(4-propylphenyl)urea 211847821037195104528574 19853461[191,192]
441-(2-fluorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea2118460510371952545285710 19853461[191,192]
451-(2-chlorophenyl)-3-(2-hydroxy-4-nitrophenyl)urea98830441037195264528570.6 19853461[191,192]
461-(2-hydroxy-4-nitrophenyl)-3-phenylurea361847210319521545285712 19853461[191,192]
471-(2-hydroxy-4-nitrophenyl)-3-(2,4,5-trichlorophenyl)urea462268151037195284528573 19853461[191,192]
481-[2-ethyl-6-(2-methylpropyl)phenyl]-3-(2-hydroxy-4-nitrophenyl)urea4622681610371952945285715 19853461[191,192]
491-(2-fluorophenyl)-3-(4-nitrophenyl)urea2985799298579945285730 19853461[191,192]
501-(2-hydroxy-4-nitrophenyl)-3-(2,4,6-trifluorophenyl)urea 4622682810371954745285715 19853461[191,192]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/cimb44100314

AMA Style

Yadav P, Lee Y-H, Panday H, Kant S, Bajwa N, Parashar R, Jha SK, Jha NK, Nand P, Lee S-S, et al. Implications of Microorganisms in Alzheimer’s Disease. Current Issues in Molecular Biology. 2022; 44(10):4584-4615. https://doi.org/10.3390/cimb44100314

Chicago/Turabian Style

Yadav, Pardeep, Yeon-Hee Lee, Hrithika Panday, Shubham Kant, Neha Bajwa, Ritika Parashar, Saurabh Kumar Jha, Niraj Kumar Jha, Parma Nand, Sang-Soo Lee, and et al. 2022. "Implications of Microorganisms in Alzheimer’s Disease" Current Issues in Molecular Biology 44, no. 10: 4584-4615. https://doi.org/10.3390/cimb44100314

Article Metrics

Back to TopTop