Skip to main content

REVIEW article

Front. Pharmacol., 28 April 2022
Sec. Experimental Pharmacology and Drug Discovery
This article is part of the Research Topic C. elegans as an Emerging Model of Pharmacological Innovation View all 6 articles

Using Caenorhabditis elegans to Model Therapeutic Interventions of Neurodegenerative Diseases Targeting Microbe-Host Interactions

  • School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China

Emerging evidence from both clinical studies and animal models indicates the importance of the interaction between the gut microbiome and the brain in the pathogenesis of neurodegenerative diseases (NDs). Although how microbes modulate neurodegeneration is still mostly unclear, recent studies have started to probe into the mechanisms for the communication between microbes and hosts in NDs. In this review, we highlight the advantages of using Caenorhabditis elegans (C. elegans) to disentangle the microbe-host interaction that regulates neurodegeneration. We summarize the microbial pro- and anti-neurodegenerative factors identified using the C. elegans ND models and the effects of many are confirmed in mouse models. Specifically, we focused on the role of bacterial amyloid proteins, such as curli, in promoting proteotoxicity and neurodegeneration by cross-seeding the aggregation of endogenous ND-related proteins, such as α-synuclein. Targeting bacterial amyloid production may serve as a novel therapeutic strategy for treating NDs, and several compounds, such as epigallocatechin-3-gallate (EGCG), were shown to suppress neurodegeneration at least partly by inhibiting curli production. Because bacterial amyloid fibrils contribute to biofilm formation, inhibition of amyloid production often leads to the disruption of biofilms. Interestingly, from a list of 59 compounds that showed neuroprotective effects in C. elegans and mouse ND models, we found that about half of them are known to inhibit bacterial growth or biofilm formation, suggesting a strong correlation between the neuroprotective and antibiofilm activities. Whether these potential therapeutics indeed protect neurons from proteotoxicity by inhibiting the cross-seeding between bacterial and human amyloid proteins awaits further investigations. Finally, we propose to screen the long list of antibiofilm agents, both FDA-approved drugs and novel compounds, for their neuroprotective effects and develop new pharmaceuticals that target the gut microbiome for the treatment of NDs. To this end, the C. elegans ND models can serve as a platform for fast, high-throughput, and low-cost drug screens that target the microbe-host interaction in NDs.

Introduction

Neurodegenerative diseases (NDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and Amyotrophic Lateral Sclerosis (ALS), are characterized at the cellular level by the aggregation of misfolded proteins into ß-sheet-rich amyloid deposits in neurons, the failure of cellular proteostasis machinery to clear out the aggregates, and mitochondrial dysfunction and energy crisis that eventually lead to neuronal death (Martin, 2012). Different types of NDs involve distinct misfolded proteins, which can not only self-propagate in a prion-like fashion but also recruit other types of proteins and convert them to misfolded conformers in a process called cross-seeding (e.g., Aβ cross-seeds the aggregation of α-synuclein) (Lim, 2019). The latter may explain the co-occurrence of multiple NDs in the same patient. Although great efforts have been devoted to developing therapeutics that can remove existing protein aggregates or prevent the formation of new ones, almost no drugs showed success in clinical trials for the treatment of NDs. Such failure suggests that our understanding of ND pathogenesis is likely incomplete, and new ideas are needed for therapeutic interventions.

One of such new ideas in the past decade came from the realization that intestinal bacteria may be a crucial predisposing factor that contributes to the development of ND through the ‘‘microbiota-gut-brain axis’’ (Peterson, 2020). ND patients have altered microbial composition in the gut compared to healthy individuals (Li C. et al., 2019). Gastrointestinal dysfunction and intestinal inflammation are positively correlated with an elevated risk of PD (Fasano et al., 2015; Chen et al., 2019), and infection with Helicobacter pylori has been associated with increased severity of PD (Tan et al., 2015; Huang HK. et al., 2018). Moreover, both clinical studies and laboratory research indicate that microbes and their metabolic products can cross the blood-brain barrier to cause chronic inflammation in the brain, which is an important risk factor for neurodegeneration in several NDs, such as AD (Cattaneo et al., 2017) and PD (Wang H. et al., 2020). Despite the emerging link between the gut microbiota and NDs, the development of therapeutics that target the microbe-host interaction in the treatment of NDs is still in its infancy, largely due to the limited mechanistic understanding of the communication between the microbiota and the host and the lack of high-throughput, physiologically relevant model systems to screen drug candidates for their therapeutic effects. In this review, we describe the use of Caenorhabditis elegans ND models to identify microbial components that affect neurodegeneration and to test chemical compounds for their potential effects in inhibiting neurodegeneration. Specifically, by closely examining the literature, we generated a comprehensive list of compounds that showed both neuroprotective effects in C. elegans ND models (oftentimes confirmed in mouse models) and inhibitory effects on bacterial biofilm formation. Our summary highlights the possibility of targeting the secretion of extracellular fibrous polymers (e.g., curli) by gut bacteria as a novel therapeutic strategy for NDs.

C. elegans ND Models

Although mouse ND models provided crucial insights into the neurodegenerative symptoms associated with neuro- and systemic inflammation caused by abnormal microbiota or pathogenic bacteria infection, the complexity of the mammalian nervous system and microbiome often makes it difficult to pinpoint the key microbial proteins or metabolites that directly impact the host neurons in the progression of neurodegeneration. Thus, the use of simpler organisms, such as C. elegans, became instrumental in disentangling the microbe-host interaction in the context of NDs.

First, C. elegans uses bacteria as their natural diet, and alteration of the bacterial genomes has been shown to affect the development and behavior of C. elegans (Watson et al., 2014). Moreover, the microbiome of C. elegans in its natural habitats has been characterized and many of these bacteria can be cultured in the laboratory (Berg et al., 2016). Second, the presence of microorganisms can be effectively controlled using a bleaching method that kills all microbes but keeps the eggs unharmed, thus allowing the cultivation of C. elegans under monoxenic conditions or with a defined mixture of microbes. Third, the nematode is transparent, so that the interaction between microbes and fluorescently labeled neurons can be visualized in live animals. Fourth, many signaling molecules and pathways are evolutionarily conserved between C. elegans and humans, indicating that the disease mechanisms found in C. elegans could be conserved in humans. Fifth, C. elegans has a short life cycle and is highly amenable to genetic manipulations. Several transgenic C. elegans ND models have been generated and provided important insights into the genetic factors that contribute to NDs. These models were also used as drug testing platforms to evaluate the therapeutic potential of various chemical compounds and natural products. Since C. elegans ND models have been extensively reviewed elsewhere (Alexander et al., 2014; Van Pelt and Truttmann, 2020), we will only briefly mention the most widely used transgenic ND models and their use in drug discovery and focus more on the microbial factors of the diseases.

A general strategy to model human NDs in C. elegans is to express the human proteins that form the protein aggregates in C. elegans muscles or neurons and to observe the degenerative phenotypes and aggregation of fluorescently labeled proteins. For AD, the first C. elegans model expressed the human Aβ peptide in the body wall muscle and found an age-dependent paralysis phenotype (Link, 1995). This model, however, has several disadvantages. First, a signal peptide is cleaved in the process of Aβ peptide generation, which leads to the production of Aβ3-42 instead of the Aβ1-42 found in human patients (McColl et al., 2012). Second, the age-dependent paralysis phenotype came under scrutiny as it is unclear whether the phenotype is a result of Aβ toxicity or intrinsic aging. To overcome these problems, other C. elegans AD models were constructed to express full-length human Aβ peptides under the control of a temperature-sensitive mRNA surveillance system that induces Aβ production after heat shock (Link et al., 2003; Hassan et al., 2009). These studies were able to observe an early-onset paralysis phenotype caused by Aβ toxicity in the muscle of young adult animals. To model the neuropathology of human AD more closely, later studies expressed Aβ in C. elegans neurons using pan-neuronal promoters and found that these AD animals have a shorter lifespan, impaired associative learning, and a significant decrease in serotonin-stimulated egg-laying (Wu et al., 2006).

The microtubule-associated protein Tau, which forms the neurofibrillary tangles in AD, was also expressed in C. elegans to model AD. The expression of the human Tau (V337M) mutants under a pan-neuronal promoter recapitulated some of the key features of AD in C. elegans, including uncoordinated movement, accumulation of insoluble tau, and age-dependent neuronal degeneration and loss. Similarly, Miyasaka et al. (2005) established the second tau model by expressing Tau mutants in the mechanosensory neurons of C. elegans; this model showed accumulation of hyperphosphorylated tau, morphological alteration of these touch neurons, and a progressive decrease in their sensory functions. More recently, Aβ and Tau co-expression models were also generated in C. elegans and showed increased deficits in associative learning, enhanced neuronal loss, and caused specific transcriptomic changes, compared to the single transgenic models (Wang et al., 2018).

Similarly, for PD models, human α-synuclein was expressed in C. elegans body wall muscles or neurons. Pan-neuronal expression of α-synuclein (A53T) mutants but not the wild-type protein caused defects in locomotion and the loss of dopaminergic neurons, which recapitulated the major aspects of PD symptoms in humans. Using the C. elegans PD model, the Caldwell group identified genetic factors that affect α-synuclein-mediated proteotoxicity via genome-wide RNAi screen and uncovered the involvement of the endocytic pathway in ameliorating α-synuclein toxicity (Hamamichi et al., 2008; Kuwahara et al., 2008).

For HD, which is caused by the polyglutamine (polyQ) expansion in the human huntingtin protein (Htt), Htt fused with polyQ repeats of different lengths were expressed in the ASH sensory neurons, which mediate avoidance behaviors to chemo- and mechanosensory stimuli. The expression of Htt-Q150 led to weak neurotoxicity in ASH neurons, but the loss of a glutamine/proline-rich protein PQE-1 significantly enhanced polyQ repeats-induced neurodegeneration (Faber et al., 2002). Using the same model, later studies found that the loss of several histone deacetylases or mutations in H3K9 methyltransferases and H3K9 methylation readers also enhanced polyQ toxicity (Bates et al., 2006; Zheng et al., 2013). Pharmacological screens with the HD model identified the neuroprotective role of mithramycin (MTR), trichostatin A (TSA), and lithium chloride (LiCl) (Voisine et al., 2007).

ALS is characterized by progressive death of motor neurons and is associated with mutations in genes encoding the Cu/Zn superoxide dismutase 1 (SOD1), RNA-binding proteins TDP-43, and fused in sarcoma (FUS). Expression of SOD1 (G85R) mutants fused with GFP in C. elegans neurons resulted in the formation of insoluble SOD1 aggregates in the perinuclear region of motor neurons and strong locomotor defects (Wang et al., 2009). Similarly, human TDP-43 (A315T) mutants were expressed in C. elegans neurons and the loss of GABAergic motor neurons and the change in locomotion speed were used as phenotyping criteria to test compounds for their neuroprotective effects against TDP-43-mediated toxicity (Boyd et al., 2014). Using the C. elegans ALS model, the Parker group screened more than 4000 FDA approved compounds and identified methylene blue, an aggregation inhibitor of the phenothiazine class, as a potent suppressor of mutant TDP-43 and FUS-induced neurotoxicity (Vaccaro et al., 2012b; Vaccaro et al., 2013; Therrien and Parker, 2014). Moreover, Kraemer and colleagues showed that inhibition of cell division cycle 7-related protein kinase (CDC7) by the small molecule inhibitor PHA767491 could reduce TDP-43 phosphorylation and prevent TDP-43-triggered neurodegeneration in C. elegans ALS models (Liachko et al., 2013).

Pro-Neurodegenerative Factors in Bacteria

Pioneering works from mouse models pointed out a pro-neurodegenerative role of the intestinal bacteria in PD animals. For example, antibiotic treatment ameliorates the pathophysiology of PD mice, and microbial recolonization after the treatment restored the PD symptoms (Sampson et al., 2016). Colonization of α-synuclein-overexpressing mice with the gut microbiota from PD patients exacerbated the physical impairments compared to transplantation of microbiota from healthy donors (Sampson et al., 2016). Metagenomic analysis of the fecal samples of PD patients revealed not only changes in gut bacterial composition (e.g., increased Lactobacillaceae and Akkermansiaceae, decreased Faecalibacterium and Roseburia (Barichella et al., 2019; Nishiwaki et al., 2020)), but also a decrease of total intestinal bacterial count compared to healthy controls (Hasegawa et al., 2015). Fecal microbiota transplantation (FMT) from healthy donors was able to alleviate the tremor and some gastrointestinal dysfunctions (e.g., constipation) in PD patients (Huang et al., 2019). Similarly, for AD, cognitive deficits, protein aggregation of Aβ and hyper-phosphorylation of tau, and synaptic plasticity were significantly improved after FMT in mouse models (Sun et al., 2019). Rapid improvement of cognitive functions in senior AD patients after FMT was reported in two independent clinical cases (Hazan, 2020; Park et al., 2021). Despite the promise, the application of FMT has its limitations due to safety concerns and the limited availability of donor microbiota. Targeted treatment of the gut microbiota in PD patients is still more desirable than the gross replacement of the microbial flora. Identification of pro-neurodegenerative factors in bacteria is the key to the development of such targeted therapy.

C. elegans ND models provide a powerful tool to systematically discover bacterial components that contribute to ND pathogenesis. Recently, using several C. elegans PD models, we screened the entire genome of E. coli to identify pro-neurodegenerative genes by feeding the E. coli single-gene knockout strains in the Keio library (Baba et al., 2006) individually to PD worms and searched for genes whose deletion led to alleviation of α-synuclein-induced locomotion defect and dopaminergic neuron death (Wang C. et al., 2021). From the 3,985 non-essential E. coli genes, we identified 38 pro-neurodegenerative genes, which fall into several genetic pathways including curli formation, lipopolysaccharide (LPS) production, lysozyme inhibition, adenosylcobalamin synthesis, oxidative stress response, metabolism, and energy homeostasis. These results suggest that a diverse array of bacteria components could promote neurodegeneration in the host.

Among the bacterial pro-neurodegenerative factors, the curli amyloid fibril has a prominent function in promoting α-synuclein aggregation through cross-seeding. Curli fibril is formed by the polymerization of the major curli subunit CsgA with the help of the membrane-bound subunit CsgB. Both CsgA and α-synuclein are enriched in ß-sheet structures, and our immunofluorescent study found that bacteria-secreted CsgA could enter C. elegans neurons and human neuroblastoma cells to seed the aggregation of α-synuclein (Wang C. et al., 2021). Although curli proteins from different bacterial species were known to cross-seed (Zhou et al., 2012), and purified CsgA was found to accelerate α-synuclein fibrilization in vitro (Sampson et al., 2020), our study provided strong evidence for in vivo cross-seeding between CsgA and α-synuclein in neurons. This cross-seeding appears to be bidirectional, since α-synuclein also facilitated the retention of CsgA in neurons. Removing csgA or csgB from the E. coli genome significantly reduced α-synuclein aggregation, rescued mitochondrial dysfunction and energy failure, and prevented the loss of dopaminergic neurons. In addition to promoting α-synuclein neurotoxicity in PD, curli also promoted the toxicity of Aβ, SOD1, and Htt-polyQ in C. elegans models of AD, ALS, and HD, respectively, likely through similar cross-seeding mechanisms (Wang C. et al., 2021). Thus, bacterial curli may have detrimental effects on a range of NDs.

The idea that amyloid proteins produced by the gut bacteria may cross-seed endogenous proteins, such as Aβ and α-synuclein, to promote neurodegeneration has been hypothesized before (Friedland, 2015) and independently validated in multiple ND models in recent studies. In addition to the C. elegans models, oral exposure to curli-producing E. coli enhanced α-synuclein deposition in the brain of aged rats (Chen et al., 2016); and colonizing germ-free mice with curli-producing E. coli exacerbated α-synuclein-induced motor impairment compared to colonization with mutant E. coli that did not produce curli (Sampson et al., 2020). Thus, the pro-neurodegenerative role of bacterial curli has been validated in multiple organisms. Targeting curli production in the gut may be a novel therapeutic approach to prevent or slow down the progression of NDs.

Besides bacterial amyloid proteins, microbial metabolites or small molecules could also promote host neurodegeneration. For example, Ray et al. (2014a), showed that an unidentified bacterial metabolite produced by Streptomyces venezuelae caused age- and dose-dependent neurodegeneration in C. elegans PD models and human SH-SY5Y neurons. This neurotoxic metabolite increased the level of ROS and damaged mitochondria, disrupted proteostasis, and enhanced the toxicity of aggregation-prone proteins in multiple C. elegans ND models (Martinez et al., 2015). Mechanistically, the metabolite acts upstream of the ubiquitin-proteasome system (UPS) and PINK (a PD-associated kinase) to regulate mitochondrial maintenance and autophagy; the well-known antioxidant glutathione (GSH) attenuated the metabolite-enhanced α-synuclein toxicity and proteasomal dysfunction (Martinez et al., 2015).

Anti-Neurodegenerative Effect of Microbes

In addition to the pro-neurodegenerative effects, studies have also found that certain bacteria and their metabolites could protect against protein aggregation and neurotoxicity. For example, the probiotic Bacillus subtilis inhibited α-synuclein aggregation and removed preformed aggregates in a C. elegans PD model (Goya et al., 2020). Interestingly, both dividing vegetative cells and environmentally resistant spores could inhibit α-synuclein aggregation but act through two distinct mechanisms: spores act via the PHA-4/Foxa dietary restriction pathways and vegetative cells via DAF-16/FOXO. Similarly, Bacillus subtilis also reduced Aβ-induced paralysis and cognitive defects and extended lifespan in a C. elegans AD model (Cogliati et al., 2020). The neuroprotective effect of B. subtilis may be mediated by beneficial gut-associated biofilm formation, the quorum-sensing peptide, and metabolites (e.g., nitric oxide). These results offer promises of using probiotics to prevent or delay neurodegeneration and suggest that altering microbial composition in the gut through nutraceutical interventions may have beneficial effects on NDs.

Some bacteria-derived compounds were shown to have anti-neurodegenerative effects. For example, mithramycin is produced by Streptomyces plicatus and is used as an antineoplastic drug to treat cancer by inhibiting RNA synthesis. Mithramycin is found to inhibit polyQ-mediated neuronal death in C. elegans HD models (Voisine et al., 2007) and to enhance motor performance and extend survival in a mouse HD model (Ferrante et al., 2004). Thus, bacteria-produced compounds, if able to cross the blood-brain barrier, may directly modulate neurodegeneration.

Microbes could also metabolize other nutrients or chemicals to produce neuroprotective effects. For example, Guo et al. (2020) found that water-soluble extracts of the herb Peganum harmala L. (wild rue) can be metabolized by E. coli OP50 (the laboratory diet for C. elegans) into oligosaccharides, which protected against polyQ-induced motility and fertility deficiency in C. elegans HD models.

Outside of the standard ND models, bacteria were also found to protect against neurotoxicity caused by leaky ion channels. Utilizing a neurotoxic allele of the mechanosensitive sodium channel to generate an ND model, Urrutia et al. (2020) found that certain bacteria species, including E. coli HT115, Comamonas aquatica, Pseudomonas aeruginosa, Stenotrophomonas humi, and Bacillus megaterium could protect neurons from leaky channel-induced degeneration. Interestingly, this neuroprotection is partially dependent on the GABA (γ-aminobutyric acid) produced by the bacteria. Since decreased GABA is associated with motor dysfunction in PD patients (Gong et al., 2018), microbe-derived GABA may also help alleviate motor defects in PD.

Neuroprotective Compounds That Inhibit Bacterial Growth and Biofilm Formation

Since bacteria can produce amyloid-forming proteins (e.g., curli produced by intestinal Enterobacteriaceae (Bian et al., 2000) and SpaP produced by Streptococcus mutans in the oral cavity (Guo et al., 2017)), which may enter neurons to cross-seed protein aggregation, one possible treatment or preventive measure of NDs would be to inhibit the production of amyloid fibril by the bacteria. To identify potential drug candidates that target this pathway, we compiled a list of 59 neuroprotective compounds that reduced neurotoxicity in C. elegans and mouse ND models and highlight the 34 compounds that also inhibited microbial growth or biofilm formation (Table 1; description of their neuroprotective effects are in Supplementary Table S1). Several compounds were also able to induce biofilm dispersal. Since the amyloid fibers are the major constituent of the extracellular matrix in biofilms, compounds that inhibit biofilm formation likely also reduce amyloid productions. Although the neuroprotective and antibiofilm effects of these compounds were mostly identified in separate studies, we attempt to make connections between these two seemingly independent effects and propose that these chemical agents may suppress neurodegeneration at least partly by inhibiting the microbial secretion of amyloid fibrils. Below, we list some examples of these potential therapeutic compounds based on their known effects on microorganisms.

TABLE 1
www.frontiersin.org

TABLE 1. Neuroprotective compounds identified in C. elegans neurodegenerative disease models and confirmed in mouse models showed effects on microorganisms.

Antibiotics

Several of the neuroprotective compounds are well-known antibiotics, including tetracycline, rifampicin, oligomycin, and bacitracin. As an example, tetracycline, the first glycylcycline antibiotic, inhibits protein synthesis by blocking the binding of aminoacyl tRNA to bacterial ribosomes and has been extensively used to treat infections of various microorganisms, including Gram-positive and Gram-negative bacteria, intracellular bacteria Chlamydiae, protozoan parasites, etc. (Chopra and Roberts, 2001). Interestingly, tetracycline was found to decrease Aβ aggregation and alleviate Aβ-induced paralysis phenotype and oxidative stress in C. elegans AD models (Diomede et al., 2010). Similarly, Balducci et al. (2018) found that long-term treatment of Doxy, a second-generation tetracycline, reduced the level of Aβ oligomers (18-mers) and significantly restored memory in a mouse AD model. Surprisingly, even an acute treatment of Doxy was sufficient to improve memory formation.

Although the exact mechanism for the neuroprotective function of antibiotics, such as tetracycline, is still unclear, it is reasonable to suspect that they suppress neurodegeneration at least partly by inhibiting bacterial growth in the gut microbiome, given the significance of the microbiota-gut-brain-axis in NDs. Therefore, the FDA-approved antibiotics can be potentially repurposed to treat NDs.

Inhibitors of Bacterial Biofilm Formation

Many of the neuroprotective compounds were found to inhibit bacterial biofilm formation, suggesting a potential link between bacterial biofilm and neurodegeneration. For example, the polyphenol epigallocatechin-3-gallate (EGCG), which is a natural compound found in green tea extract, has been well-known for its effects in reducing oxidative stress, inhibiting protein aggregation, and protecting against neurodegeneration in PD and AD (Singh et al., 2016). At the same time, EGCG also has broad-spectrum effects in inhibiting biofilm formation (Serra et al., 2016). It is, however, unclear whether these two functions are connected.

Our recent study disentangled these two functions by feeding PD C. elegans with bacteria pre-treated with EGCG (Wang C. et al., 2021). In this scenario, only the bacteria but not the neurons are treated by EGCG. We found that EGCG strongly inhibits curli production and biofilm formation in E. coli bacteria. Importantly, treating the bacteria alone with EGCG provides strong protection against α-synuclein-induced neurodegeneration, which is almost indistinguishable from the effects of treating both the bacteria and the PD animals. Therefore, the neuroprotective effects of EGCG may be largely due to its activities in inhibiting the curli expression and bacterial biofilm formation.

Another example came from the bioactive components of Ginkgo biloba extract. G. biloba has an extensive history of being used to treat dementia in traditional Chinese medicine. In C. elegans AD models, G. biloba extract and one of its components, ginkgolide A, was found to reduce Aβ oligomerization and deposition and inhibit Aβ-induced paralysis and chemotaxis defects (Wu et al., 2006). In a mouse AD model, the same extract also reduced Aβ toxicity, improved cognitive functions, and induced neurogenesis in the hippocampus (Tchantchou et al., 2007). These studies support the use of G. biloba extracts as neuroprotective agents. Interestingly, G. biloba extracts and ginkgolic acid could block biofilm formation in E. coli O157:H7, Staphylococcus aureus, Salmonella and Listeria and downregulate the expression of curli structural subunit csgA in E. coli K12 (Lee et al., 2014; Wu et al., 2016). Thus, just like EGCG, the natural products in Ginkgo biloba extract may also exert neuroprotective effects by inhibiting curli production in gut bacteria.

From our literature search, we found 27 compounds that inhibit both neurodegeneration and bacterial biofilm formation (compounds with asterisks in Table 1). The correlation between the two activities in these compounds deserves further investigation. We hypothesize that at least some of these compounds may suppress neurodegeneration by blocking the cross-seeding of the bacterial amyloid proteins with ND-associated aggregation-prone proteins. Nevertheless, we could not rule out the possibility that some compounds may exert neuroprotective effects through multiple mechanisms that also include the inhibition of ER stress and oxidative stress (see below).

Inhibitors of Fungal Biofilm Formation

Among the neuroprotective agents, a few have antifungal effects and could inhibit fungal biofilm formation. For example, clioquinol is an antifungal drug wildly used to treat skin infections such as infected eczema and athlete’s foot. Clioquinol inhibits Candida albicans biofilm formation in a dose-dependent manner by disrupting metal ion homeostasis (You et al., 2020). Unexpectedly, clioquinol was also found to promote the degradation of Aβ oligomers and rescue Aβ toxicity in a C. elegans AD model (Matlack et al., 2014). Similarly, clioquinol could reduce Aβ burden and reverse memory impairment in a mouse AD model (Grossi et al., 2009). These studies highlight the possibility, although not tested, that the neuroprotective effects of clioquinol may be connected to its activity in regulating metal ion metabolism and biofilm formation in the microbes.

Bacterial Biofilm, ER Stress, and Oxidative Stress in NDs

At the cellular level, the mechanisms of neurological damage in NDs involve protein aggregation, mitochondrial dysfunction, oxidative stress, calcium homeostasis dysfunction, and neuroinflammation (Jellinger, 2010). The loss of cellular homeostasis often leads to the activation of the endoplasmic reticulum (ER) stress-triggered unfolded protein response (UPR) pathway and the impairment of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, which play vital roles in ND pathogenesis (Branca et al., 2017; Ren et al., 2021).

ER stress is induced by disturbances in the structure and function of the ER with the accumulation of misfolded proteins and alterations in the calcium homeostasis. For example, tau aggregates trigger abnormal interactions between ER proteins and the essential components of ER-associated degradation (ERAD) in AD brains, leading to ER stress (Meier et al., 2015). Conversely, overexpression of xbp-1, a major regulator of UPR, alleviated ER stress and protected dopaminergic neurons from α-synuclein-induced neurotoxicity (Ray et al., 2014b). Intriguingly, pathogenic bacterial biofilm was also found to induce host ER stress. For example, when forming host-associated biofilms, Group A Streptococcus (GAS), a human pathogen that causes a range of infections, could secrete streptolysins, which induce host ER-stress in both mammalian cells and an in vivo mouse model (Vajjala et al., 2019). Thus, inhibiting microbial biofilm formation may reduce ER stress and provide beneficial effects for neurons in ND patients. Indeed, many of the neuroprotective compounds (e.g., Salubrinal in Table 1) showed activities of both inhibiting biofilm formation and reducing ER stress.

Nrf2-ARE pathway, an indicator and regulator of oxidative stress, plays an important role in protecting neurons from degeneration in many NDs. Reduced Nrf2 levels were found in human AD and PD brains and in animal models of AD (Branca et al., 2017; Ramsey et al., 2007). Removing Nrf2 increased the levels of Aβ and phosphorylated tau and enhanced neurodegeneration in a mouse AD model (Branca et al., 2017; Rojo et al., 2018), whereas activating Nrf2 (by knocking down its negative regulator) led to the reduction in oxidative stress and neuroinflammation (Williamson et al., 2012). Several compounds in our list (e.g., metformin and caffeine in Table 1) were shown to pharmacologically activate Nrf2, induce the expression of antioxidant enzymes, and protect neurons against degeneration (Link et al., 2003; Dostal et al., 2010; Boettler et al., 2011; Cui et al., 2016; Saewanee et al., 2021). For example, in a C. elegans model of AD, caffeine induced the nuclear translocation of SKN-1 (the C. elegans homolog of Nrf2) and delayed Aβ-mediated paralysis (Dostal et al., 2010). Given that these compounds also inhibit the formation of bacterial biofilms, it is unclear whether they suppress neurodegeneration by inhibiting cross-seeding or inducing antioxidative response or both. Reduced protein aggregation by the inhibition of cross-seeding may also facilitate the activation of the antioxidant Nrf2-ARE pathway.

Oxidative stress often exacerbates ER stress in NDs. During oxidative stress, the accumulation of reactive oxygen species (ROS) disrupts the redox-dependent protein folding process and thus increases the production of misfolded proteins, which further enhance ER stress and proteotoxicity in neurons. Alleviating both ER stress and oxidative stress provide synergistic benefit for the treatment of NDs. For example, curcumin, a polyphenol compound from the curry spice turmeric, possesses potent antioxidant and anti-UPR activities and could modulate multiple targets implicated in the pathogenesis of NDs (Lim et al., 2001; Begum et al., 2008). In fact, curcumin was shown to alleviate Aβ and tau-induced neurotoxicity and protein aggregation in C. elegans AD models (Alavez et al., 2011; Miyasaka et al., 2016).

Interestingly, curcumin shows anti-bacterial activity against a variety of infections when administrated together with antibiotics (Kali et al., 2016). Curcumin can inhibit biofilm formation, perturb bacterial membranes, disturb bacterial cell division, and alter gene expression patterns (Vaughn et al., 2017). Thus, although it is unclear whether the neuroprotective effect of curcumin relates to its bactericidal activity, this example raises the possibility of targeting bacterial biofilm to simultaneously reduce both ER stress and oxidative stress in NDs.

Discussion and Future Perspectives

The gut microbiome holds the promise of becoming the therapeutic target of NDs, which currently have no effective treatments. Understanding the molecular mechanisms by which intestinal bacteria modulate neurodegeneration is, however, challenging, given the complexity of the microbial composition in the gut and the difficulties of studying the effects of a single bacterial component in isolation in a well-controlled system. Therefore, the use of simple model organisms like C. elegans can provide unparalleled advantages in studying the communication between microbes and neurons in the context of NDs. As we have shown above, using a variety of C. elegans ND models, both pro- and anti-neurodegenerative factors can be identified from the bacteria, paving the way for a mechanistic understanding of how bacterial proteins and metabolites affect host neurodegeneration.

Nevertheless, the C. elegans ND models also have certain limitations compared to rodent models. For example, C. elegans lacks the complex immune system found in mammals. Although certain molecular pathways in innate immunity are conserved between C. elegans and humans (Ermolaeva and Schumacher, 2014), C. elegans has no specialized immune cells, no adaptive immunity, and no typical inflammatory response. Thus, it will be difficult to use C. elegans models to investigate the effects of the microbes in triggering neuroinflammation. Interestingly, C. elegans does have glia cells comparable to mammalian microglia, but their roles in neurodegeneration have not been studied. The absence of a complex immune system in C. elegans ND models, however, simplifies the studies of microbe-neuron interaction and allows direct molecular interaction to be revealed.

In this review, we paid specific attention to bacterial amyloid proteins and biofilm formation as an important pro-neurodegenerative mechanism in microbes, given the cross-seeding between bacterial amyloid proteins (e.g., curli) and human endogenous aggregation-prone proteins (e.g., α-synuclein), both of which are enriched in ß-sheet structures. Guided by this cross-seeding mechanism, we searched the literature to identify compounds that showed both neuroprotective effects in ND models and antibiofilm effects on microorganisms and raised the hypothesis that some of these therapeutic compounds may suppress neurodegeneration at least partly through inhibiting bacterial amyloid production (which leads to antibiofilm activities) and thus preventing cross-seeding.

Although direct evidence demonstrating the causal relationship between the antibiofilm and neuroprotective activities are still missing for most compounds except for a few (e.g., EGCG (Wang C. et al., 2021), 30 (51%) out of the 59 neuroprotective compounds we found have known effects of inhibiting bacterial growth or biofilm formation, suggesting that the correlation of these two activities is quite strong. The percentage may be even higher, given that the effects on microorganisms are not tested for many of these compounds. Although the list we compiled (Table 1) is in no way a complete list, we hope it could inspire fellow researchers to consider the alteration of gut microbiota as a possible pharmacological mechanism of neuroprotective agents or to develop drugs that specifically target the intestinal microbes for treating NDs.

In fact, previous works have identified a wide range of anti-biofilm agents including FDA-approved drugs (Gilbert-Girard et al., 2020) and novel compounds (Junker and Clardy, 2007; Paytubi et al., 2017). It will be of great interest to test their neuroprotective effects with the attempts of repurposing them for the treatment of human NDs in future research. Again, the C. elegans ND models could be instrumental for screening these compounds for potential anti-neurodegenerative activities, given the convenience of setting up fast and high-throughput drug screens using C. elegans. Moving forward, with a deeper understanding of the mechanisms underlying the microbiota-gut-brain interactions in NDs and more therapeutic candidates targeting the gut microbiome for ND treatment, we expect a potential paradigm shift in the research of ND pathogenesis and drug development.

Author Contributions

CW and CZ wrote the draft and edited it. CW prepared the Table. CZ secured the funding and supervised the study. Both authors read and approved the manuscript.

Funding

This work is supported by grants from the Food and Health Bureau of Hong Kong (HMRF 07183186 to CZ), the Research Grants Council of Hong Kong (ECS 27104219, GRF 17107021, and CRF C7026-20G to CZ), the University of Hong Kong (seed fund 201910159087 and 202011159053 to CZ), and the National Natural Science Foundation of China (Excellent Young Scientists Fund for Hong Kong and Macau 32122002 to CZ).

Conflict of Interest

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

Publisher’s Note

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

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphar.2022.875349/full#supplementary-material

References

Abbas, H. A., Elsherbini, A. M., and Shaldam, M. A. (2017). Repurposing Metformin as a Quorum Sensing Inhibitor in Pseudomonas aeruginosa. Afr. Health Sci. 17, 808–819. doi:10.4314/ahs.v17i3.24

PubMed Abstract | CrossRef Full Text | Google Scholar

Abbas, S., and Wink, M. (2010). Epigallocatechin Gallate Inhibits Beta Amyloid Oligomerization in Caenorhabditis elegans and Affects the Daf-2/insulin-like Signaling Pathway. Phytomedicine 17, 902–909. doi:10.1016/j.phymed.2010.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Abidi, S. H., Ahmed, K., and Kazmi, S. U. (2019). The Antibiofilm Activity of Acetylsalicylic Acid, Mefenamic Acid, Acetaminophen against Biofilms Formed by P. aeruginosa and S. Epidermidis. J. Pak Med. Assoc. 69, 1493–1495. doi:10.5455/jpma.295488

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahmad, W., and Ebert, P. R. (2017). Metformin Attenuates Aβ Pathology Mediated through Levamisole Sensitive Nicotinic Acetylcholine Receptors in a C. elegans Model of Alzheimer's Disease. Mol. Neurobiol. 54, 5427–5439. doi:10.1007/s12035-016-0085-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Alavez, S., Vantipalli, M. C., Zucker, D. J., Klang, I. M., and Lithgow, G. J. (2011). Amyloid-binding Compounds Maintain Protein Homeostasis during Ageing and Extend Lifespan. Nature 472, 226–229. doi:10.1038/nature09873

PubMed Abstract | CrossRef Full Text | Google Scholar

Alexander, A. G., Marfil, V., and Li, C. (2014). Use of Caenorhabditis elegans as a Model to Study Alzheimer's Disease and Other Neurodegenerative Diseases. Front. Genet. 5, 279. doi:10.3389/fgene.2014.00279

PubMed Abstract | CrossRef Full Text | Google Scholar

Arendash, G. W., Schleif, W., Rezai-Zadeh, K., Jackson, E. K., Zacharia, L. C., Cracchiolo, J. R., et al. (2006). Caffeine Protects Alzheimer's Mice against Cognitive Impairment and Reduces Brain Beta-Amyloid Production. Neuroscience 142, 941–952. doi:10.1016/j.neuroscience.2006.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Ay, M., Luo, J., Langley, M., Jin, H., Anantharam, V., Kanthasamy, A., et al. (2017). Molecular Mechanisms Underlying Protective Effects of Quercetin against Mitochondrial Dysfunction and Progressive Dopaminergic Neurodegeneration in Cell Culture and MitoPark Transgenic Mouse Models of Parkinson's Disease. J. Neurochem. 141, 766–782. doi:10.1111/jnc.14033

PubMed Abstract | CrossRef Full Text | Google Scholar

Baba, T., Ara, T., Hasegawa, M., Takai, Y., Okumura, Y., Baba, M., et al. (2006). Construction of Escherichia coli K-12 In-Frame, Single-Gene Knockout Mutants: the Keio Collection. Mol. Syst. Biol. 2, 2006–0008. doi:10.1038/msb4100050

PubMed Abstract | CrossRef Full Text | Google Scholar

Balducci, C., Santamaria, G., La Vitola, P., Brandi, E., Grandi, F., Viscomi, A. R., et al. (2018). Doxycycline Counteracts Neuroinflammation Restoring Memory in Alzheimer's Disease Mouse Models. Neurobiol. Aging 70, 128–139. doi:10.1016/j.neurobiolaging.2018.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Barichella, M., Severgnini, M., Cilia, R., Cassani, E., Bolliri, C., Caronni, S., et al. (2019). Unraveling Gut Microbiota in Parkinson's Disease and Atypical Parkinsonism. Mov Disord. 34, 396–405. doi:10.1002/mds.27581

PubMed Abstract | CrossRef Full Text | Google Scholar

Bates, E. A., Victor, M., Jones, A. K., Shi, Y., and Hart, A. C. (2006). Differential Contributions of Caenorhabditis elegans Histone Deacetylases to Huntingtin Polyglutamine Toxicity. J. Neurosci. 26, 2830–2838. doi:10.1523/JNEUROSCI.3344-05.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Begum, A. N., Jones, M. R., Lim, G. P., Morihara, T., Kim, P., Heath, D. D., et al. (2008). Curcumin Structure-Function, Bioavailability, and Efficacy in Models of Neuroinflammation and Alzheimer's Disease. J. Pharmacol. Exp. Ther. 326, 196–208. doi:10.1124/jpet.108.137455

PubMed Abstract | CrossRef Full Text | Google Scholar

Berg, M., Stenuit, B., Ho, J., Wang, A., Parke, C., Knight, M., et al. (2016). Assembly of the Caenorhabditis elegans Gut Microbiota from Diverse Soil Microbial Environments. ISME J. 10, 1998–2009. doi:10.1038/ismej.2015.253

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhattacharya, S., Haertel, C., Maelicke, A., and Montag, D. (2014). Galantamine Slows Down Plaque Formation and Behavioral Decline in the 5XFAD Mouse Model of Alzheimer's Disease. Plos One 9, e89454. doi:10.1371/journal.pone.0089454

PubMed Abstract | CrossRef Full Text | Google Scholar

Bian, Z., Brauner, A., Li, Y., and Normark, S. (2000). Expression of and Cytokine Activation by Escherichia coli Curli Fibers in Human Sepsis. J. Infect. Dis. 181, 602–612. doi:10.1086/315233

PubMed Abstract | CrossRef Full Text | Google Scholar

Boettler, U., Sommerfeld, K., Volz, N., Pahlke, G., Teller, N., Somoza, V., et al. (2011). Coffee Constituents as Modulators of Nrf2 Nuclear Translocation and ARE (EpRE)-dependent Gene Expression. J. Nutr. Biochem. 22, 426–440. doi:10.1016/j.jnutbio.2010.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Bondia, P., Flors, C., and Torra, J. (2021). Boosting the Inactivation of Bacterial Biofilms by Photodynamic Targeting of Matrix Structures with Thioflavin T. Chem. Commun. 57, 8648–8651. doi:10.1039/d1cc03155d

CrossRef Full Text | Google Scholar

Borges, A., Saavedra, M. J., and Simões, M. (2012). The Activity of Ferulic and Gallic Acids in Biofilm Prevention and Control of Pathogenic Bacteria. Biofouling 28, 755–767. doi:10.1080/08927014.2012.706751

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyd, J. D., Lee, P., Feiler, M. S., Zauur, N., Liu, M., Concannon, J., et al. (2014). A High-Content Screen Identifies Novel Compounds that Inhibit Stress-Induced TDP-43 Cellular Aggregation and Associated Cytotoxicity. J. Biomol. Screen. 19, 44–56. doi:10.1177/1087057113501553

PubMed Abstract | CrossRef Full Text | Google Scholar

Branca, C., Ferreira, E., Nguyen, T. V., Doyle, K., Caccamo, A., and Oddo, S. (2017). Genetic Reduction of Nrf2 Exacerbates Cognitive Deficits in a Mouse Model of Alzheimer's Disease. Hum. Mol. Genet. 26, 4823–4835. doi:10.1093/hmg/ddx361

PubMed Abstract | CrossRef Full Text | Google Scholar

Braungart, E., Gerlach, M., Riederer, P., Baumeister, R., and Hoener, M. C. (2004). Caenorhabditis elegans MPP+ Model of Parkinson's Disease for High-Throughput Drug Screenings. Neurodegener Dis. 1, 175–183. doi:10.1159/000080983

PubMed Abstract | CrossRef Full Text | Google Scholar

Büttner, S., Broeskamp, F., Sommer, C., Markaki, M., Habernig, L., Alavian-Ghavanini, A., et al. (2014). Spermidine Protects against α-synuclein Neurotoxicity. Cell Cycle 13, 3903–3908. doi:10.4161/15384101.2014.973309

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, W. J., Huang, J. H., Zhang, S. Q., Wu, B., Kapahi, P., Zhang, X. M., et al. (2011). Icariin and its Derivative Icariside II Extend Healthspan via insulin/IGF-1 Pathway in C. elegans. Plos One 6, e28835. doi:10.1371/journal.pone.0028835

PubMed Abstract | CrossRef Full Text | Google Scholar

Calamini, B., Silva, M. C., Madoux, F., Hutt, D. M., Khanna, S., Chalfant, M. A., et al. (2010). “ML346: A Novel Modulator of Proteostasis for Protein Conformational Diseases,” in Probe Reports from the NIH Molecular Libraries Program.

Google Scholar

Cao, X., Ye, Q., Fan, M., and Liu, C. (2019). Antimicrobial Effects of the Ginsenoside Rh2 on Monospecies and Multispecies Cariogenic Biofilms. J. Appl. Microbiol. 126, 740–751. doi:10.1111/jam.14178

PubMed Abstract | CrossRef Full Text | Google Scholar

Cattaneo, A., Cattane, N., Galluzzi, S., Provasi, S., Lopizzo, N., Festari, C., et al. (2017). Association of Brain Amyloidosis with Pro-inflammatory Gut Bacterial Taxa and Peripheral Inflammation Markers in Cognitively Impaired Elderly. Neurobiol. Aging 49, 60–68. doi:10.1016/j.neurobiolaging.2016.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Chakraborty, P., Dastidar, D. G., Paul, P., Dutta, S., Basu, D., Sharma, S. R., et al. (2020). Inhibition of Biofilm Formation of Pseudomonas aeruginosa by Caffeine: a Potential Approach for Sustainable Management of Biofilm. Arch. Microbiol. 202, 623–635. doi:10.1007/s00203-019-01775-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Chalorak, P., Sanguanphun, T., Limboonreung, T., and Meemon, K. (2021). Neurorescue Effects of Frondoside A and Ginsenoside Rg3 in C. elegans Model of Parkinson's Disease. Molecules 26. doi:10.3390/molecules26164843

CrossRef Full Text | Google Scholar

Chan, S., Kantham, S., Rao, V. M., Palanivelu, M. K., Pham, H. L., Shaw, P. N., et al. (2016). Metal Chelation, Radical Scavenging and Inhibition of Aβ₄₂ Fibrillation by Food Constituents in Relation to Alzheimer's Disease. Food Chem. 199, 185–194. doi:10.1016/j.foodchem.2015.11.118

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Chen, Y., and Pu, J. (2018). Leucine-Rich Repeat Kinase 2 in Parkinson's Disease: Updated from Pathogenesis to Potential Therapeutic Target. Eur. Neurol. 79, 256–265. doi:10.1159/000488938

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, K. S., Menezes, K., Rodgers, J. B., O'Hara, D. M., Tran, N., Fujisawa, K., et al. (2021). Small Molecule Inhibitors of α-synuclein Oligomers Identified by Targeting Early Dopamine-Mediated Motor Impairment in C. elegans. Mol. Neurodegener 16, 77. doi:10.1186/s13024-021-00497-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Q. Q., Haikal, C., Li, W., and Li, J. Y. (2019). Gut Inflammation in Association with Pathogenesis of Parkinson's Disease. Front. Mol. Neurosci. 12, 218. doi:10.3389/fnmol.2019.00218

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, S. G., Stribinskis, V., Rane, M. J., Demuth, D. R., Gozal, E., Roberts, A. M., et al. (2016). Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis elegans. Sci. Rep. 6, 34477. doi:10.1038/srep34477

PubMed Abstract | CrossRef Full Text | Google Scholar

Cherny, R. A., Ayton, S., Finkelstein, D. I., Bush, A. I., McColl, G., and Massa, S. M. (2012). PBT2 Reduces Toxicity in a C. elegans Model of polyQ Aggregation and Extends Lifespan, Reduces Striatal Atrophy and Improves Motor Performance in the R6/2 Mouse Model of Huntington's Disease. J. Huntingtons Dis. 1, 211–219. doi:10.3233/JHD-120029

PubMed Abstract | CrossRef Full Text | Google Scholar

Chin, P. C., Liu, L., Morrison, B. E., Siddiq, A., Ratan, R. R., Bottiglieri, T., et al. (2004). The C-Raf Inhibitor GW5074 Provides Neuroprotection In Vitro and in an Animal Model of Neurodegeneration through a MEK-ERK and Akt-independent Mechanism. J. Neurochem. 90, 595–608. doi:10.1111/j.1471-4159.2004.02530.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ching, T. T., Chiang, W. C., Chen, C. S., and Hsu, A. L. (2011). Celecoxib Extends C. elegans Lifespan via Inhibition of Insulin-like Signaling but Not Cyclooxygenase-2 Activity. Aging Cell 10, 506–519. doi:10.1111/j.1474-9726.2011.00688.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Chiu, C. T., Liu, G., Leeds, P., and Chuang, D. M. (2011). Combined Treatment with the Mood Stabilizers Lithium and Valproate Produces Multiple Beneficial Effects in Transgenic Mouse Models of Huntington's Disease. Neuropsychopharmacology 36, 2406–2421. doi:10.1038/npp.2011.128

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, N., Kim, H. W., Lee, H. K., Jeon, B. J., and Sung, S. H. (2016). Ameliorative Effect of Betulin from Betula Platyphylla Bark on Scopolamine-Induced Amnesic Mice. Biosci. Biotechnol. Biochem. 80, 166–171. doi:10.1080/09168451.2015.1072460

PubMed Abstract | CrossRef Full Text | Google Scholar

Chopra, I., and Roberts, M. (2001). Tetracycline Antibiotics: Mode of Action, Applications, Molecular Biology, and Epidemiology of Bacterial Resistance. Microbiol. Mol. Biol. Rev. 65, 232–contents. doi:10.1128/MMBR.65.2.232-260.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung, Y. H., Lin, C. W., Huang, H. Y., Chen, S. L., Huang, H. J., Sun, Y. C., et al. (2020). Targeting Inflammation, PHA-767491 Shows a Broad Spectrum in Protein Aggregation Diseases. J. Mol. Neurosci. 70, 1140–1152. doi:10.1007/s12031-020-01521-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Coenye, T., Brackman, G., Rigole, P., De Witte, E., Honraet, K., Rossel, B., et al. (2012). Eradication of Propionibacterium Acnes Biofilms by Plant Extracts and Putative Identification of Icariin, Resveratrol and Salidroside as Active Compounds. Phytomedicine 19, 409–412. doi:10.1016/j.phymed.2011.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Cogliati, S., Clementi, V., Francisco, M., Crespo, C., Argañaraz, F., and Grau, R. (2020). Bacillus Subtilis Delays Neurodegeneration and Behavioral Impairment in the Alzheimer's Disease Model Caenorhabditis Elegans. J. Alzheimers Dis. 73, 1035–1052. doi:10.3233/JAD-190837

PubMed Abstract | CrossRef Full Text | Google Scholar

Crowe, A., Henderson, M. J., Anderson, J., Titus, S. A., Zakharov, A., Simeonov, A., et al. (2020). Compound Screening in Cell-Based Models of Tau Inclusion Formation: Comparison of Primary Neuron and HEK293 Cell Assays. J. Biol. Chem. 295, 4001–4013. doi:10.1074/jbc.RA119.010532

PubMed Abstract | CrossRef Full Text | Google Scholar

Cuevas, E., Burks, S., Raymick, J., Robinson, B., Gómez-Crisóstomo, N. P., Escudero-Lourdes, C., et al. (2020). Tauroursodeoxycholic Acid (TUDCA) Is Neuroprotective in a Chronic Mouse Model of Parkinson's Disease. Nutr. Neurosci. 1, 1–18. doi:10.1080/1028415X.2020.1859729

CrossRef Full Text | Google Scholar

Cui, Q., Li, X., and Zhu, H. (2016). Curcumin Ameliorates Dopaminergic Neuronal Oxidative Damage via Activation of the Akt/Nrf2 Pathway. Mol. Med. Rep. 13, 1381–1388. doi:10.3892/mmr.2015.4657

PubMed Abstract | CrossRef Full Text | Google Scholar

Cui, W. Q., Qu, Q. W., Wang, J. P., Bai, J. W., Bello-Onaghise, G., Li, Y. A., et al. (2019). Discovery of Potential Anti-infective Therapy Targeting Glutamine Synthetase in Staphylococcus Xylosus. Front. Chem. 7, 381. doi:10.3389/fchem.2019.00381

PubMed Abstract | CrossRef Full Text | Google Scholar

Dasagrandhi, C., Park, S., Jung, W. K., and Kim, Y. M. (2018). Antibacterial and Biofilm Modulating Potential of Ferulic Acid-Grafted Chitosan against Human Pathogenic Bacteria. Int. J. Mol. Sci. 19. doi:10.3390/ijms19082157

PubMed Abstract | CrossRef Full Text | Google Scholar

De Jesús-Cortés, H., Xu, P., Drawbridge, J., Estill, S. J., Huntington, P., Tran, S., et al. (2012). Neuroprotective Efficacy of Aminopropyl Carbazoles in a Mouse Model of Parkinson Disease. Proc. Natl. Acad. Sci. U S A. 109, 17010–17015. doi:10.1073/pnas.1213956109

PubMed Abstract | CrossRef Full Text | Google Scholar

De Oliveira, D. M. P., Bohlmann, L., Conroy, T., Jen, F. E., Everest-Dass, A., Hansford, K. A., et al. (2020). Repurposing a Neurodegenerative Disease Drug to Treat Gram-Negative Antibiotic-Resistant Bacterial Sepsis. Sci. Transl Med. 12. doi:10.1126/scitranslmed.abb3791

CrossRef Full Text | Google Scholar

Ding, T., Li, T., Wang, Z., and Li, J. (2017). Curcumin Liposomes Interfere with Quorum Sensing System of Aeromonas Sobria and In Silico Analysis. Sci. Rep. 7, 8612. doi:10.1038/s41598-017-08986-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Diomede, L., Cassata, G., Fiordaliso, F., Salio, M., Ami, D., Natalello, A., et al. (2010). Tetracycline and its Analogues Protect Caenorhabditis elegans from β Amyloid-Induced Toxicity by Targeting Oligomers. Neurobiol. Dis. 40, 424–431. doi:10.1016/j.nbd.2010.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, G., Liu, H., Yu, X., Zhang, X., Lu, H., Zhou, T., et al. (2018). Antimicrobial and Anti-biofilm Activity of Tannic Acid against Staphylococcus aureus. Nat. Prod. Res. 32, 2225–2228. doi:10.1080/14786419.2017.1366485

PubMed Abstract | CrossRef Full Text | Google Scholar

Dostal, V., Roberts, C. M., and Link, C. D. (2010). Genetic Mechanisms of Coffee Extract protection in a Caenorhabditis elegans Model of β-amyloid Peptide Toxicity. Genetics 186, 857–866. doi:10.1534/genetics.110.120436

PubMed Abstract | CrossRef Full Text | Google Scholar

Doub, J. B., Heil, E. L., Ntem-Mensah, A., Neeley, R., and Ching, P. R. (2020). Rifabutin Use in Staphylococcus Biofilm Infections: A Case Series. Antibiotics 9, 326. doi:10.3390/antibiotics9060326

PubMed Abstract | CrossRef Full Text | Google Scholar

Dragicevic, N., Smith, A., Lin, X., Yuan, F., Copes, N., Delic, V., et al. (2011). Green tea Epigallocatechin-3-Gallate (EGCG) and Other Flavonoids Reduce Alzheimer's Amyloid-Induced Mitochondrial Dysfunction. J. Alzheimers Dis. 26, 507–521. doi:10.3233/JAD-2011-101629

PubMed Abstract | CrossRef Full Text | Google Scholar

Ermolaeva, M. A., and Schumacher, B. (2014). Insights from the Worm: the C. elegans Model for Innate Immunity. Semin. Immunol. 26, 303–309. doi:10.1016/j.smim.2014.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Eskelinen, M. H., and Kivipelto, M. (2010). Caffeine as a Protective Factor in Dementia and Alzheimer's Disease. J. Alzheimers Dis. 20 Suppl 1, S167–S174. doi:10.3233/Jad-2010-1404

PubMed Abstract | CrossRef Full Text | Google Scholar

Evason, K., Collins, J. J., Huang, C., Hughes, S., and Kornfeld, K. (2008). Valproic Acid Extends Caenorhabditis elegans Lifespan. Aging Cell 7, 305–317. doi:10.1111/j.1474-9726.2008.00375.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Faber, P. W., Voisine, C., King, D. C., Bates, E. A., and Hart, A. C. (2002). Glutamine/proline-rich PQE-1 Proteins Protect Caenorhabditis elegans Neurons from Huntingtin Polyglutamine Neurotoxicity. Proc. Natl. Acad. Sci. U S A. 99, 17131–17136. doi:10.1073/pnas.262544899

PubMed Abstract | CrossRef Full Text | Google Scholar

Farr, S. A., Roesler, E., Niehoff, M. L., Roby, D. A., McKee, A., and Morley, J. E. (2019). Metformin Improves Learning and Memory in the SAMP8 Mouse Model of Alzheimer's Disease. J. Alzheimers Dis. 68, 1699–1710. doi:10.3233/JAD-181240

PubMed Abstract | CrossRef Full Text | Google Scholar

Fasano, A., Visanji, N. P., Liu, L. W., Lang, A. E., and Pfeiffer, R. F. (2015). Gastrointestinal Dysfunction in Parkinson's Disease. Lancet Neurol. 14, 625–639. doi:10.1016/S1474-4422(15)00007-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Fatouros, C., Pir, G. J., Biernat, J., Koushika, S. P., Mandelkow, E., Mandelkow, E. M., et al. (2012). Inhibition of Tau Aggregation in a Novel Caenorhabditis elegans Model of Tauopathy Mitigates Proteotoxicity. Hum. Mol. Genet. 21, 3587–3603. doi:10.1093/hmg/dds190

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferrante, R. J., Ryu, H., Kubilus, J. K., D'Mello, S., Sugars, K. L., Lee, J., et al. (2004). Chemotherapy for the Brain: the Antitumor Antibiotic Mithramycin Prolongs Survival in a Mouse Model of Huntington's Disease. J. Neurosci. 24, 10335–10342. doi:10.1523/JNEUROSCI.2599-04.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Friedland, R. P. (2015). Mechanisms of Molecular Mimicry Involving the Microbiota in Neurodegeneration. J. Alzheimers Dis. 45, 349–362. doi:10.3233/JAD-142841

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, R. H., Wang, Y. C., Chen, C. S., Tsai, R. T., Liu, S. P., Chang, W. L., et al. (2014). Acetylcorynoline Attenuates Dopaminergic Neuron Degeneration and α-synuclein Aggregation in Animal Models of Parkinson's Disease. Neuropharmacology 82, 108–120. doi:10.1016/j.neuropharm.2013.08.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Fung, T. C., Vuong, H. E., Luna, C. D. G., Pronovost, G. N., Aleksandrova, A. A., Riley, N. G., et al. (2019). Intestinal Serotonin and Fluoxetine Exposure Modulate Bacterial Colonization in the Gut. Nat. Microbiol. 4, 2064–2073. doi:10.1038/s41564-019-0540-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Gamir-Morralla, A., Sacristán, S., Medina, M., and Iglesias, T. (2019). Effects of Thioflavin T and GSK-3 Inhibition on Lifespan and Motility in a Caenorhabditis elegans Model of Tauopathy. J. Alzheimers Dis. Rep. 3, 47–57. doi:10.3233/ADR-180087

PubMed Abstract | CrossRef Full Text | Google Scholar

Garnaud, C., Champleboux, M., Maubon, D., and Govin, M. J. (2016). Histone Deacetylases and Their Inhibition in Candida Species. Front. Microbiol. 7, 1238. doi:10.3389/fmicb.2016.01238

PubMed Abstract | CrossRef Full Text | Google Scholar

Gilbert-Girard, S., Savijoki, K., Yli-Kauhaluoma, J., and Fallarero, A. (2020). Screening of FDA-Approved Drugs Using a 384-Well Plate-Based Biofilm Platform: The Case of Fingolimod. Microorganisms 8. doi:10.3390/microorganisms8111834

PubMed Abstract | CrossRef Full Text | Google Scholar

Gong, T., Xiang, Y., Saleh, M. G., Gao, F., Chen, W., Edden, R. A. E., et al. (2018). Inhibitory Motor Dysfunction in Parkinson's Disease Subtypes. J. Magn. Reson. Imaging 47, 1610–1615. doi:10.1002/jmri.25865

PubMed Abstract | CrossRef Full Text | Google Scholar

Goya, M. E., Xue, F., Sampedro-Torres-Quevedo, C., Arnaouteli, S., Riquelme-Dominguez, L., Romanowski, A., et al. (2020). Probiotic Bacillus Subtilis Protects against α-Synuclein Aggregation in C. elegans. Cell Rep 30, 367–e7. e367. doi:10.1016/j.celrep.2019.12.078

PubMed Abstract | CrossRef Full Text | Google Scholar

Grossi, C., Francese, S., Casini, A., Rosi, M. C., Luccarini, I., Fiorentini, A., et al. (2009). Clioquinol Decreases Amyloid-Beta burden and Reduces Working Memory Impairment in a Transgenic Mouse Model of Alzheimer's Disease. J. Alzheimers Dis. 17, 423–440. doi:10.3233/JAD-2009-1063

PubMed Abstract | CrossRef Full Text | Google Scholar

Gu, C., Hu, Q., Wu, J., Mu, C., Ren, H., Liu, C. F., et al. (2018). P7C3 Inhibits LPS-Induced Microglial Activation to Protect Dopaminergic Neurons against Inflammatory Factor-Induced Cell Death In Vitro and In Vivo. Front Cel Neurosci 12, 400. doi:10.3389/fncel.2018.00400

PubMed Abstract | CrossRef Full Text | Google Scholar

Guan, X.-N. Z. T., Zhang, T., Yang, T., Dong, Z., Yang, S., Lan, L., et al. (2022). Covalent Sortase A Inhibitor ML346 Prevents Staphylococcus aureus Infection of Galleria Mellonella. RSC Med. Chem. 13, 138–149. doi:10.1039/d1md00316j

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, L., Shokeen, B., He, X., Shi, W., and Lux, R. (2017). Streptococcus Mutans SpaP Binds to RadD of Fusobacterium Nucleatum Ssp. Polymorphum. Mol. Oral Microbiol. 32, 355–364. doi:10.1111/omi.12177

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, X., Yuan, J., Song, X., Wang, X., Sun, Q., Tian, J., et al. (2020). Bacteria Metabolites from Peganum Harmala L. Polysaccharides Inhibits polyQ Aggregation through Proteasome-Mediated Protein Degradation in C. elegans. Int. J. Biol. Macromol 161, 681–691. doi:10.1016/j.ijbiomac.2020.06.091

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutierrez-Zepeda, A., Santell, R., Wu, Z., Brown, M., Wu, Y., Khan, I., et al. (2005). Soy Isoflavone Glycitein Protects against Beta Amyloid-Induced Toxicity and Oxidative Stress in Transgenic Caenorhabditis elegans. BMC Neurosci. 6, 54. doi:10.1186/1471-2202-6-54

PubMed Abstract | CrossRef Full Text | Google Scholar

Halliday, M., Radford, H., Zents, K. A. M., Molloy, C., Moreno, J. A., Verity, N. C., et al. (2017). Repurposed Drugs Targeting eIF2α-P-Mediated Translational Repression Prevent Neurodegeneration in Mice. Brain 140, 1768–1783. doi:10.1093/brain/awx074

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamamichi, S., Rivas, R. N., Knight, A. L., Cao, S., Caldwell, K. A., and Caldwell, G. A. (2008). Hypothesis-based RNAi Screening Identifies Neuroprotective Genes in a Parkinson's Disease Model. Proc. Natl. Acad. Sci. U S A. 105, 728–733. doi:10.1073/pnas.0711018105

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasegawa, S., Goto, S., Tsuji, H., Okuno, T., Asahara, T., Nomoto, K., et al. (2015). Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson's Disease. Plos One 10, e0142164. doi:10.1371/journal.pone.0142164

PubMed Abstract | CrossRef Full Text | Google Scholar

Hassan, W. M., Merin, D. A., Fonte, V., and Link, C. D. (2009). AIP-1 Ameliorates Beta-Amyloid Peptide Toxicity in a Caenorhabditis elegans Alzheimer's Disease Model. Hum. Mol. Genet. 18, 2739–2747. doi:10.1093/hmg/ddp209

PubMed Abstract | CrossRef Full Text | Google Scholar

Hazan, S. (2020). Rapid Improvement in Alzheimer's Disease Symptoms Following Fecal Microbiota Transplantation: a Case Report. J. Int. Med. Res. 48, 300060520925930. doi:10.1177/0300060520925930

PubMed Abstract | CrossRef Full Text | Google Scholar

Himeno, E., Ohyagi, Y., Ma, L., Nakamura, N., Miyoshi, K., Sakae, N., et al. (2011). Apomorphine Treatment in Alzheimer Mice Promoting Amyloid-β Degradation. Ann. Neurol. 69, 248–256. doi:10.1002/ana.22319

PubMed Abstract | CrossRef Full Text | Google Scholar

Hobley, L., Li, B., Wood, J. L., Kim, S. H., Naidoo, J., Ferreira, A. S., et al. (2017). Spermidine Promotes Bacillus Subtilis Biofilm Formation by Activating Expression of the Matrix Regulator slrR. J. Biol. Chem. 292, 12041–12053. doi:10.1074/jbc.M117.789644

PubMed Abstract | CrossRef Full Text | Google Scholar

Hosokawa, M., Arai, T., Masuda-Suzukake, M., Nonaka, T., Yamashita, M., Akiyama, H., et al. (2012). Methylene Blue Reduced Abnormal Tau Accumulation in P301L Tau Transgenic Mice. Plos One 7, e52389. doi:10.1371/journal.pone.0052389

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, H., Xu, H., Luo, Q., He, J., Li, M., Chen, H., et al. (2019). Fecal Microbiota Transplantation to Treat Parkinson's Disease with Constipation: A Case Report. Medicine (Baltimore) 98, e16163. doi:10.1097/MD.0000000000016163

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, H. K., Wang, J. H., Lei, W. Y., Chen, C. L., Chang, C. Y., and Liou, L. S. (2018a). Helicobacter pylori Infection Is Associated with an Increased Risk of Parkinson's Disease: A Population-Based Retrospective Cohort Study. Parkinsonism Relat. Disord. 47, 26–31. doi:10.1016/j.parkreldis.2017.11.331

CrossRef Full Text | Google Scholar

Huang, M., Liang, Y., Chen, H., Xu, B., Chai, C., and Xing, P. (2018b). The Role of Fluoxetine in Activating Wnt/β-Catenin Signaling and Repressing β-Amyloid Production in an Alzheimer Mouse Model. Front. Aging Neurosci. 10, 164. doi:10.3389/fnagi.2018.00164

PubMed Abstract | CrossRef Full Text | Google Scholar

Inden, M., Kitamura, Y., Abe, M., Tamaki, A., Takata, K., and Taniguchi, T. (2011). Parkinsonian Rotenone Mouse Model: Reevaluation of Long-Term Administration of Rotenone in C57BL/6 Mice. Biol. Pharm. Bull. 34, 92–96. doi:10.1248/bpb.34.92

PubMed Abstract | CrossRef Full Text | Google Scholar

Jellinger, K. A. (2010). Basic Mechanisms of Neurodegeneration: a Critical Update. J. Cel Mol Med 14, 457–487. doi:10.1111/j.1582-4934.2010.01010.x

CrossRef Full Text | Google Scholar

Junker, L. M., and Clardy, J. (2007). High-throughput Screens for Small-Molecule Inhibitors of Pseudomonas aeruginosa Biofilm Development. Antimicrob. Agents Chemother. 51, 3582–3590. doi:10.1128/AAC.00506-07

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaizaki, A., Tien, L. T., Pang, Y., Cai, Z., Tanaka, S., Numazawa, S., et al. (2013). Celecoxib Reduces Brain Dopaminergic Neuronaldysfunction, and Improves Sensorimotor Behavioral Performance in Neonatal Rats Exposed to Systemic Lipopolysaccharide. J. Neuroinflammation 10, 45. doi:10.1186/1742-2094-10-45

PubMed Abstract | CrossRef Full Text | Google Scholar

Kali, A., Bhuvaneshwar, D., Charles, P. M., and Seetha, K. S. (2016). Antibacterial Synergy of Curcumin with Antibiotics against Biofilm Producing Clinical Bacterial Isolates. J. Basic Clin. Pharm. 7, 93–96. doi:10.4103/0976-0105.183265

PubMed Abstract | CrossRef Full Text | Google Scholar

Kautu, B. B., Carrasquilla, A., Hicks, M. L., Caldwell, K. A., and Caldwell, G. A. (2013). Valproic Acid Ameliorates C. elegans Dopaminergic Neurodegeneration with Implications for ERK-MAPK Signaling. Neurosci. Lett. 541, 116–119. doi:10.1016/j.neulet.2013.02.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Keowkase, R., Aboukhatwa, M., Adam, B. L., Beach, J. W., Terry, A. V., Buccafussco, J. J., et al. (2010a). Neuroprotective Effects and Mechanism of Cognitive-Enhancing Choline Analogs JWB 1-84-1 and JAY 2-22-33 in Neuronal Culture and Caenorhabditis elegans. Mol. Neurodegener 5, 59. doi:10.1186/1750-1326-5-59

PubMed Abstract | CrossRef Full Text | Google Scholar

Keowkase, R., Aboukhatwa, M., and Luo, Y. (2010b). Fluoxetine Protects against Amyloid-Beta Toxicity, in Part via Daf-16 Mediated Cell Signaling Pathway, in Caenorhabditis elegans. Neuropharmacology 59, 358–365. doi:10.1016/j.neuropharm.2010.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Kidd, S. K., and Schneider, J. S. (2011). Protective Effects of Valproic Acid on the Nigrostriatal Dopamine System in a 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model of Parkinson's Disease. Neuroscience 194, 189–194. doi:10.1016/j.neuroscience.2011.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Koh, S. H., Lee, S. M., Kim, H. Y., Lee, K. Y., Lee, Y. J., Kim, H. T., et al. (2006). The Effect of Epigallocatechin Gallate on Suppressing Disease Progression of ALS Model Mice. Neurosci. Lett. 395, 103–107. doi:10.1016/j.neulet.2005.10.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Kosuru, R. Y., Roy, A., and Bera, S. (2021). Antagonistic Roles of Gallates and Ascorbic Acid in Pyomelanin Biosynthesis of Pseudomonas aeruginosa Biofilms. Curr. Microbiol. 78, 3843–3852. doi:10.1007/s00284-021-02655-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Koutzoumis, D. N., Vergara, M., Pino, J., Buddendorff, J., Khoshbouei, H., Mandel, R. J., et al. (2020). Alterations of the Gut Microbiota with Antibiotics Protects Dopamine Neuron Loss and Improve Motor Deficits in a Pharmacological Rodent Model of Parkinson's Disease. Exp. Neurol. 325, 113159. doi:10.1016/j.expneurol.2019.113159

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurniawan, A., and Yamamoto, T. (2013). Biofilm Polymer for Biosorption of Pollutant Ions. Proced. Environ. Sci. 17, 179–187. doi:10.1016/j.proenv.2013.02.027

CrossRef Full Text | Google Scholar

Kuwahara, T., Koyama, A., Koyama, S., Yoshina, S., Ren, C. H., Kato, T., et al. (2008). A Systematic RNAi Screen Reveals Involvement of Endocytic Pathway in Neuronal Dysfunction in Alpha-Synuclein Transgenic C. elegans. Hum. Mol. Genet. 17, 2997–3009. doi:10.1093/hmg/ddn198

PubMed Abstract | CrossRef Full Text | Google Scholar

Lalouckova, K., Mala, L., Marsik, P., and Skrivanova, E. (2021). In Vitro Antibacterial Effect of the Methanolic Extract of the Korean Soybean Fermented Product Doenjang against Staphylococcus aureus. Animals 11, 2319. doi:10.3390/ani11082319

PubMed Abstract | CrossRef Full Text | Google Scholar

Laloux, C., Derambure, P., Houdayer, E., Jacquesson, J. M., Bordet, R., Destée, A., et al. (2008). Effect of Dopaminergic Substances on Sleep/wakefulness in saline- and MPTP-Treated Mice. J. Sleep Res. 17, 101–110. doi:10.1111/j.1365-2869.2008.00625.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, J. H., Kim, Y. G., Ryu, S. Y., Cho, M. H., and Lee, J. (2014). Ginkgolic Acids and Ginkgo Biloba Extract Inhibit Escherichia coli O157:H7 and Staphylococcus aureus Biofilm Formation. Int. J. Food Microbiol. 174, 47–55. doi:10.1016/j.ijfoodmicro.2013.12.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Lehtonen, Š., Jaronen, M., Vehviläinen, P., Lakso, M., Rudgalvyte, M., Keksa-Goldsteine, V., et al. (2016). Inhibition of Excessive Oxidative Protein Folding Is Protective in MPP(+) Toxicity-Induced Parkinson's Disease Models. Antioxid. Redox Signal. 25, 485–497. doi:10.1089/ars.2015.6402

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Cui, L., Yang, Y., Miao, J., Zhao, X., Zhang, J., et al. (2019a). Gut Microbiota Differs between Parkinson's Disease Patients and Healthy Controls in Northeast China. Front. Mol. Neurosci. 12, 171. doi:10.3389/fnmol.2019.00171

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, F., Zhang, Y., Lu, X., Shi, J., and Gong, Q. (2019b). Icariin Improves the Cognitive Function of APP/PS1 Mice via Suppressing Endoplasmic Reticulum Stress. Life Sci. 234, 116739. doi:10.1016/j.lfs.2019.116739

PubMed Abstract | CrossRef Full Text | Google Scholar

Liachko, N. F., McMillan, P. J., Guthrie, C. R., Bird, T. D., Leverenz, J. B., and Kraemer, B. C. (2013). CDC7 Inhibition Blocks Pathological TDP-43 Phosphorylation and Neurodegeneration. Ann. Neurol. 74, 39–52. doi:10.1002/ana.23870

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, G. P., Chu, T., Yang, F., Beech, W., Frautschy, S. A., and Cole, G. M. (2001). The Curry Spice Curcumin Reduces Oxidative Damage and Amyloid Pathology in an Alzheimer Transgenic Mouse. J. Neurosci. 21, 8370–8377. doi:10.1523/jneurosci.21-21-08370.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, K. H. (2019). Diverse Misfolded Conformational Strains and Cross-Seeding of Misfolded Proteins Implicated in Neurodegenerative Diseases. Front. Mol. Neurosci. 12, 158. doi:10.3389/fnmol.2019.00158

PubMed Abstract | CrossRef Full Text | Google Scholar

Link, C. D. (1995). Expression of Human Beta-Amyloid Peptide in Transgenic Caenorhabditis elegans. Proc. Natl. Acad. Sci. U S A. 92, 9368–9372. doi:10.1073/pnas.92.20.9368

PubMed Abstract | CrossRef Full Text | Google Scholar

Link, C. D., Taft, A., Kapulkin, V., Duke, K., Kim, S., Fei, Q., et al. (2003). Gene Expression Analysis in a Transgenic Caenorhabditis elegans Alzheimer's Disease Model. Neurobiol. Aging 24, 397–413. doi:10.1016/s0197-4580(02)00224-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Hamamichi, S., Lee, B. D., Yang, D., Ray, A., Caldwell, G. A., et al. (2011). Inhibitors of LRRK2 Kinase Attenuate Neurodegeneration and Parkinson-like Phenotypes in Caenorhabditis elegans and Drosophila Parkinson's Disease Models. Hum. Mol. Genet. 20, 3933–3942. doi:10.1093/hmg/ddr312

PubMed Abstract | CrossRef Full Text | Google Scholar

Locke, C. J., Fox, S. A., Caldwell, G. A., and Caldwell, K. A. (2008). Acetaminophen Attenuates Dopamine Neuron Degeneration in Animal Models of Parkinson's Disease. Neurosci. Lett. 439, 129–133. doi:10.1016/j.neulet.2008.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Lublin, A., Isoda, F., Patel, H., Yen, K., Nguyen, L., Hajje, D., et al. (2011). FDA-approved Drugs that Protect Mammalian Neurons from Glucose Toxicity Slow Aging Dependent on Cbp and Protect against Proteotoxicity. Plos One 6, ARTN e27762. doi:10.1371/journal.pone.0027762

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, J., Dong, B., Wang, K., Cai, S., Liu, T., Cheng, X., et al. (2017). Baicalin Inhibits Biofilm Formation, Attenuates the Quorum Sensing-Controlled Virulence and Enhances Pseudomonas aeruginosa Clearance in a Mouse Peritoneal Implant Infection Model. Plos One 12, e0176883. doi:10.1371/journal.pone.0176883

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, J., Wang, R., Chen, T., Jiang, S., and Xu, A. (2021). Protective Effects of Baicalin in a Caenorhabditis elegans Model of Parkinson's Disease. Toxicol. Res. (Camb) 10, 409–417. doi:10.1093/toxres/tfaa107

PubMed Abstract | CrossRef Full Text | Google Scholar

Martin, L. J. (2012). Biology of Mitochondria in Neurodegenerative Diseases. Prog. Mol. Biol. Transl Sci. 107, 355–415. doi:10.1016/B978-0-12-385883-2.00005-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinez, B. A., Kim, H., Ray, A., Caldwell, G. A., and Caldwell, K. A. (2015). A Bacterial Metabolite Induces Glutathione-Tractable Proteostatic Damage, Proteasomal Disturbances, and PINK1-dependent Autophagy in C. elegans. Cell Death Dis 6, e1908. doi:10.1038/cddis.2015.270

PubMed Abstract | CrossRef Full Text | Google Scholar

Matlack, K. E. S., Tardiff, D. F., Narayan, P., Hamamichi, S., Caldwell, K. A., Caldwell, G. A., et al. (2014). Clioquinol Promotes the Degradation of Metal-dependent Amyloid-β (Aβ) Oligomers to Restore Endocytosis and Ameliorate Aβ Toxicity. Proc. Natl. Acad. Sci. U.S.A. 111, 4013–4018. doi:10.1073/pnas.1402228111

PubMed Abstract | CrossRef Full Text | Google Scholar

McColl, G., Roberts, B. R., Pukala, T. L., Kenche, V. B., Roberts, C. M., Link, C. D., et al. (2012). Utility of an Improved Model of Amyloid-Beta (Aβ₁₋₄₂) Toxicity in Caenorhabditis elegans for Drug Screening for Alzheimer's Disease. Mol. Neurodegener 7, 57. doi:10.1186/1750-1326-7-57

PubMed Abstract | CrossRef Full Text | Google Scholar

McCormick, A. V., Wheeler, J. M., Guthrie, C. R., Liachko, N. F., and Kraemer, B. C. (2013). Dopamine D2 Receptor Antagonism Suppresses Tau Aggregation and Neurotoxicity. Biol. Psychiatry 73, 464–471. doi:10.1016/j.biopsych.2012.08.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Meier, S., Bell, M., Lyons, D. N., Ingram, A., Chen, J., Gensel, J. C., et al. (2015). Identification of Novel Tau Interactions with Endoplasmic Reticulum Proteins in Alzheimer's Disease Brain. J. Alzheimers Dis. 48, 687–702. doi:10.3233/JAD-150298

PubMed Abstract | CrossRef Full Text | Google Scholar

Memariani, H., Memariani, M., and Ghasemian, A. (2019). An Overview on Anti-biofilm Properties of Quercetin against Bacterial Pathogens. World J. Microbiol. Biotechnol. 35, 143. doi:10.1007/s11274-019-2719-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyasaka, T., Ding, Z., Gengyo-Ando, K., Oue, M., Yamaguchi, H., Mitani, S., et al. (2005). Progressive Neurodegeneration in C. elegans Model of Tauopathy. Neurobiol. Dis. 20, 372–383. doi:10.1016/j.nbd.2005.03.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyasaka, T., Xie, C., Yoshimura, S., Shinzaki, Y., Yoshina, S., Kage-Nakadai, E., et al. (2016). Curcumin Improves Tau-Induced Neuronal Dysfunction of Nematodes. Neurobiol. Aging 39, 69–81. doi:10.1016/j.neurobiolaging.2015.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Mocko, J. B., Kern, A., Moosmann, B., Behl, C., and Hajieva, P. (2010). Phenothiazines Interfere with Dopaminergic Neurodegeneration in Caenorhabditis elegans Models of Parkinson's Disease. Neurobiol. Dis. 40, 120–129. doi:10.1016/j.nbd.2010.03.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Mori, T., Rezai-Zadeh, K., Koyama, N., Arendash, G. W., Yamaguchi, H., Kakuda, N., et al. (2012). Tannic Acid Is a Natural β-secretase Inhibitor that Prevents Cognitive Impairment and Mitigates Alzheimer-like Pathology in Transgenic Mice. J. Biol. Chem. 287, 6912–6927. doi:10.1074/jbc.M111.294025

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishiwaki, H., Ito, M., Ishida, T., Hamaguchi, T., Maeda, T., Kashihara, K., et al. (2020). Meta-Analysis of Gut Dysbiosis in Parkinson's Disease. Mov Disord. 35, 1626–1635. doi:10.1002/mds.28119

PubMed Abstract | CrossRef Full Text | Google Scholar

Ogawa, N., Tanaka, K., Asanuma, M., Kawai, M., Masumizu, T., Kohno, M., et al. (1994). Bromocriptine Protects Mice against 6-hydroxydopamine and Scavenges Hydroxyl Free Radicals In Vitro. Brain Res. 657, 207–213. doi:10.1016/0006-8993(94)90969-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Opoku-Temeng, C., Miller, N. J., and Sintim, H. O. (2017). Hydroxybenzylidene-indolinones, C-Di-AMP Synthase Inhibitors, Have Antibacterial and Anti-biofilm Activities and Also Re-sensitize Resistant Bacteria to Methicillin and Vancomycin. RSC Adv. 7, 8288–8294. doi:10.1039/c6ra28443d

CrossRef Full Text | Google Scholar

Park, S. H., Lee, J. H., Shin, J., Kim, J. S., Cha, B., Lee, S., et al. (2021). Cognitive Function Improvement after Fecal Microbiota Transplantation in Alzheimer's Dementia Patient: a Case Report. Curr. Med. Res. Opin. 37, 1739–1744. doi:10.1080/03007995.2021.1957807

PubMed Abstract | CrossRef Full Text | Google Scholar

Patil, S. P., Jain, P. D., Ghumatkar, P. J., Tambe, R., and Sathaye, S. (2014). Neuroprotective Effect of Metformin in MPTP-Induced Parkinson's Disease in Mice. Neuroscience 277, 747–754. doi:10.1016/j.neuroscience.2014.07.046

PubMed Abstract | CrossRef Full Text | Google Scholar

Paytubi, S., de La Cruz, M., Tormo, J. R., Martín, J., González, I., González-Menendez, V., et al. (2017). A High-Throughput Screening Platform of Microbial Natural Products for the Discovery of Molecules with Antibiofilm Properties against Salmonella. Front. Microbiol. 8, 326. doi:10.3389/fmicb.2017.00326

PubMed Abstract | CrossRef Full Text | Google Scholar

Pelling, H., Nzakizwanayo, J., Milo, S., Denham, E. L., MacFarlane, W. M., Bock, L. J., et al. (2019). Bacterial Biofilm Formation on Indwelling Urethral Catheters. Lett. Appl. Microbiol. 68, 277–293. doi:10.1111/lam.13144

PubMed Abstract | CrossRef Full Text | Google Scholar

Peterson, C. T. (2020). Dysfunction of the Microbiota-Gut-Brain Axis in Neurodegenerative Disease: The Promise of Therapeutic Modulation with Prebiotics, Medicinal Herbs, Probiotics, and Synbiotics. J. Evid. Based Integr. Med. 25, 2515690X20957225. doi:10.1177/2515690X20957225

PubMed Abstract | CrossRef Full Text | Google Scholar

Pham, J. V., Yilma, M. A., Feliz, A., Majid, M. T., Maffetone, N., Walker, J. R., et al. (2019). A Review of the Microbial Production of Bioactive Natural Products and Biologics. Front. Microbiol. 10, 1404. doi:10.3389/fmicb.2019.01404

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramsey, C. P., Glass, C. A., Montgomery, M. B., Lindl, K. A., Ritson, G. P., Chia, L. A., et al. (2007). Expression of Nrf2 in Neurodegenerative Diseases. J. Neuropathol. Exp. Neurol. 66, 75–85. doi:10.1097/nen.0b013e31802d6da9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ray, A., Martinez, B. A., Berkowitz, L. A., Caldwell, G. A., and Caldwell, K. A. (2014a). Mitochondrial Dysfunction, Oxidative Stress, and Neurodegeneration Elicited by a Bacterial Metabolite in a C. elegans Parkinson's Model. Cel Death Dis 5, e984. doi:10.1038/cddis.2013.513

CrossRef Full Text | Google Scholar

Ray, A., Zhang, S., Rentas, C., Caldwell, K. A., and Caldwell, G. A. (2014b). RTCB-1 Mediates Neuroprotection via XBP-1 mRNA Splicing in the Unfolded Protein Response Pathway. J. Neurosci. 34, 16076–16085. doi:10.1523/JNEUROSCI.1945-14.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

Regitz, C., Dussling, L. M., and Wenzel, U. (2014). Amyloid-beta (Aβ₁₋₄₂)-Induced Paralysis in Caenorhabditis elegans Is Inhibited by the Polyphenol Quercetin through Activation of Protein Degradation Pathways. Mol. Nutr. Food Res. 58, 1931–1940. doi:10.1002/mnfr.201400014

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, H., Zhai, W., Lu, X., and Wang, G. (2021). The Cross-Links of Endoplasmic Reticulum Stress, Autophagy, and Neurodegeneration in Parkinson's Disease. Front. Aging Neurosci. 13, 691881. doi:10.3389/fnagi.2021.691881

PubMed Abstract | CrossRef Full Text | Google Scholar

Rojas, F., Gonzalez, D., Cortes, N., Ampuero, E., Hernández, D. E., Fritz, E., et al. (2015). Reactive Oxygen Species Trigger Motoneuron Death in Non-cell-autonomous Models of ALS through Activation of C-Abl Signaling. Front. Cel Neurosci 9, 203. doi:10.3389/fncel.2015.00203

PubMed Abstract | CrossRef Full Text | Google Scholar

Rojo, A. I., Pajares, M., García-Yagüe, A. J., Buendia, I., Van Leuven, F., Yamamoto, M., et al. (2018). Deficiency in the Transcription Factor NRF2 Worsens Inflammatory Parameters in a Mouse Model with Combined Tauopathy and Amyloidopathy. Redox Biol. 18, 173–180. doi:10.1016/j.redox.2018.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Saewanee, N., Praputpittaya, T., Malaiwong, N., Chalorak, P., and Meemon, K. (2021). Neuroprotective Effect of Metformin on Dopaminergic Neurodegeneration and α-synuclein Aggregation in C. elegans Model of Parkinson's Disease. Neurosci. Res. 162, 13–21. doi:10.1016/j.neures.2019.12.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Sampson, T. R., Challis, C., Jain, N., Moiseyenko, A., Ladinsky, M. S., Shastri, G. G., et al. (2020). A Gut Bacterial Amyloid Promotes α-synuclein Aggregation and Motor Impairment in Mice. Elife 9. doi:10.7554/eLife.53111

CrossRef Full Text | Google Scholar

Sampson, T. R., Debelius, J. W., Thron, T., Janssen, S., Shastri, G. G., Ilhan, Z. E., et al. (2016). Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson's Disease. Cell 167, 1469–e12. doi:10.1016/j.cell.2016.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchis, A., García-Gimeno, M. A., Cañada-Martínez, A. J., Sequedo, M. D., Millán, J. M., Sanz, P., et al. (2019). Metformin Treatment Reduces Motor and Neuropsychiatric Phenotypes in the zQ175 Mouse Model of Huntington Disease. Exp. Mol. Med. 51, 1–16. doi:10.1038/s12276-019-0264-9

CrossRef Full Text | Google Scholar

Sandlie, I., Solberg, K., and Kleppe, K. (1980). The Effect of Caffeine on Cell Growth and Metabolism of Thymidine in Escherichia coli. Mutat. Res. 73, 29–41. doi:10.1016/0027-5107(80)90133-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, S., Raymick, J., Ray, B., Lahiri, D. K., Paule, M. G., and Schmued, L. (2015). Oral Administration of Thioflavin T Prevents Beta Amyloid Plaque Formation in Double Transgenic AD Mice. Curr. Alzheimer Res. 12, 837–846. doi:10.2174/156720501209151019105647

PubMed Abstract | CrossRef Full Text | Google Scholar

Saxena, S., Cabuy, E., and Caroni, P. (2009). A Role for Motoneuron Subtype-Selective ER Stress in Disease Manifestations of FALS Mice. Nat. Neurosci. 12, 627–636. doi:10.1038/nn.2297

PubMed Abstract | CrossRef Full Text | Google Scholar

Sedjahtera, A., Gunawan, L., Bray, L., Hung, L. W., Parsons, J., Okamura, N., et al. (2018). Targeting Metals Rescues the Phenotype in an Animal Model of Tauopathy. Metallomics 10, 1339–1347. doi:10.1039/c8mt00153g

PubMed Abstract | CrossRef Full Text | Google Scholar

Serra, D. O., Mika, F., Richter, A. M., and Hengge, R. (2016). The green tea Polyphenol EGCG Inhibits E. coli Biofilm Formation by Impairing Amyloid Curli Fibre Assembly and Downregulating the Biofilm Regulator CsgD via the σ(E) -dependent sRNA RybB. Mol. Microbiol. 101, 136–151. doi:10.1111/mmi.13379

PubMed Abstract | CrossRef Full Text | Google Scholar

Shaw, J. D., Brodke, D. S., Williams, D. L., and Ashton, N. N. (2020). Methylene Blue Is an Effective Disclosing Agent for Identifying Bacterial Biofilms on Orthopaedic Implants. J. Bone Jt. Surg Am 102, 1784–1791. doi:10.2106/JBJS.20.00091

PubMed Abstract | CrossRef Full Text | Google Scholar

Siddiqui, M. F., Winters, H., Maqbool, F., Qayyum, S., Singh, L., Ullah, I., et al. (2019). Tannic Acid Treatment to Deter Microbial Biofouling in Flow Cell System and on RO Membrane in Drip Flow Reactor. Dwt 171, 62–66. doi:10.5004/dwt.2019.24767

CrossRef Full Text | Google Scholar

Siles, S. A., Srinivasan, A., Pierce, C. G., Lopez-Ribot, J. L., and Ramasubramanian, A. K. (2013). High-throughput Screening of a Collection of Known Pharmacologically Active Small Compounds for Identification of Candida Albicans Biofilm Inhibitors. Antimicrob. Agents Chemother. 57, 3681–3687. doi:10.1128/AAC.00680-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, D., Gupta, S., Verma, I., Morsy, M. A., Nair, A. B., and Ahmed, A. F. (2021). Hidden Pharmacological Activities of Valproic Acid: A New Insight. Biomed. Pharmacother. 142, 112021. doi:10.1016/j.biopha.2021.112021

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, N. A., Mandal, A. K., and Khan, Z. A. (2016). Potential Neuroprotective Properties of Epigallocatechin-3-Gallate (EGCG). Nutr. J. 15, 60. doi:10.1186/s12937-016-0179-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Sood, A., Warren Beach, J., Webster, S. J., Terry, A. V., and Buccafusco, J. J. (2007). The Effects of JWB1-84-1 on Memory-Related Task Performance by Amyloid Abeta Transgenic Mice and by Young and Aged Monkeys. Neuropharmacology 53, 588–600. doi:10.1016/j.neuropharm.2007.06.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Stone, G., Wood, P., Dixon, L., Keyhan, M., and Matin, A. (2002). Tetracycline Rapidly Reaches All the Constituent Cells of Uropathogenic Escherichia coli Biofilms. Antimicrob. Agents Chemother. 46, 2458–2461. doi:10.1128/AAC.46.8.2458-2461.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, J., Xu, J., Ling, Y., Wang, F., Gong, T., Yang, C., et al. (2019). Fecal Microbiota Transplantation Alleviated Alzheimer's Disease-like Pathogenesis in APP/PS1 Transgenic Mice. Transl Psychiatry 9, 189. doi:10.1038/s41398-019-0525-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Suo, H., Wang, P., Tong, J., Cai, L., Liu, J., Huang, D., et al. (2015). NRSF Is an Essential Mediator for the Neuroprotection of Trichostatin A in the MPTP Mouse Model of Parkinson's Disease. Neuropharmacology 99, 67–78. doi:10.1016/j.neuropharm.2015.07.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Suresh, D., Sabir, S., Yu, T. T., Wenholz, D., Das, T., Black, D. S., et al. (2021). Natural Product Rottlerin Derivatives Targeting Quorum Sensing. Molecules 26. doi:10.3390/molecules26123745

CrossRef Full Text | Google Scholar

Takahashi, H., Kashimura, M., Koiso, H., Kuda, T., and Kimura, B. (2013). Use of Ferulic Acid as a Novel Candidate of Growth Inhibiting Agent against Listeria Monocytogenes in Ready-To-Eat Food. Food Control 33, 244–248. doi:10.1016/j.foodcont.2013.03.013

CrossRef Full Text | Google Scholar

Tan, A. H., Mahadeva, S., Marras, C., Thalha, A. M., Kiew, C. K., Yeat, C. M., et al. (2015). Helicobacter pylori Infection Is Associated with Worse Severity of Parkinson's Disease. Parkinsonism Relat. Disord. 21, 221–225. doi:10.1016/j.parkreldis.2014.12.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Tauffenberger, A., Julien, C., and Parker, J. A. (2013). Evaluation of Longevity Enhancing Compounds against Transactive Response DNA-Binding Protein-43 Neuronal Toxicity. Neurobiol. Aging 34, 2175–2182. doi:10.1016/j.neurobiolaging.2013.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Tchantchou, F., Xu, Y., Wu, Y., Christen, Y., and Luo, Y. (2007). EGb 761 Enhances Adult Hippocampal Neurogenesis and Phosphorylation of CREB in Transgenic Mouse Model of Alzheimer's Disease. FASEB J. 21, 2400–2408. doi:10.1096/fj.06-7649com

PubMed Abstract | CrossRef Full Text | Google Scholar

Therrien, M., and Parker, J. A. (2014). Worming Forward: Amyotrophic Lateral Sclerosis Toxicity Mechanisms and Genetic Interactions in Caenorhabditis elegans. Front. Genet. 5, 85. doi:10.3389/fgene.2014.00085

PubMed Abstract | CrossRef Full Text | Google Scholar

Thongbhubate, K., Nakafuji, Y., Matsuoka, R., Kakegawa, S., and Suzuki, H. (2021). Effect of Spermidine on Biofilm Formation in Escherichia coli K-12. J. Bacteriol. 203. doi:10.1128/JB.00652-20

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsai, C. W., Tsai, R. T., Liu, S. P., Chen, C. S., Tsai, M. C., Chien, S. H., et al. (2017). Neuroprotective Effects of Betulin in Pharmacological and Transgenic Caenorhabditis elegans Models of Parkinson's Disease. Cel Transpl. 26, 1903–1918. doi:10.1177/0963689717738785

PubMed Abstract | CrossRef Full Text | Google Scholar

Tzeng, S. R., Huang, Y. W., Zhang, Y. Q., Yang, C. Y., Chien, H. S., Chen, Y. R., et al. (2020). A Celecoxib Derivative Eradicates Antibiotic-Resistant Staphylococcus aureus and Biofilms by Targeting YidC2 Translocase. Int. J. Mol. Sci. 21. doi:10.3390/ijms21239312

PubMed Abstract | CrossRef Full Text | Google Scholar

Umeda, T., Tanaka, A., Sakai, A., Yamamoto, A., Sakane, T., and Tomiyama, T. (2018). Intranasal Rifampicin for Alzheimer's Disease Prevention. Alzheimers Dement (N Y) 4, 304–313. doi:10.1016/j.trci.2018.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Urrutia, A., García-Angulo, V. A., Fuentes, A., Caneo, M., Legüe, M., Urquiza, S., et al. (2020). Bacterially Produced Metabolites Protect C. elegans Neurons from Degeneration. Plos Biol. 18, e3000638. doi:10.1371/journal.pbio.3000638

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaccaro, A., Patten, S. A., Aggad, D., Julien, C., Maios, C., Kabashi, E., et al. (2013). Pharmacological Reduction of ER Stress Protects against TDP-43 Neuronal Toxicity In Vivo. Neurobiol. Dis. 55, 64–75. doi:10.1016/j.nbd.2013.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaccaro, A., Patten, S. A., Ciura, S., Maios, C., Therrien, M., Drapeau, P., et al. (2012a). Methylene Blue Protects against TDP-43 and FUS Neuronal Toxicity in C. elegans and D. rerio. Plos One 7, e42117. doi:10.1371/journal.pone.0042117

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaccaro, A., Tauffenberger, A., Ash, P. E., Carlomagno, Y., Petrucelli, L., and Parker, J. A. (2012b). TDP-1/TDP-43 Regulates Stress Signaling and Age-dependent Proteotoxicity in Caenorhabditis elegans. Plos Genet. 8, e1002806. doi:10.1371/journal.pgen.1002806

PubMed Abstract | CrossRef Full Text | Google Scholar

Vajjala, A., Biswas, D., Tay, W. H., Hanski, E., and Kline, K. A. (2019). Streptolysin-induced Endoplasmic Reticulum Stress Promotes Group A Streptococcal Host-Associated Biofilm Formation and Necrotising Fasciitis. Cell Microbiol 21, e12956. doi:10.1111/cmi.12956

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Pelt, K. M., and Truttmann, M. C. (2020). Caenorhabditis elegans as a Model System for Studying Aging-Associated Neurodegenerative Diseases. Transl Med. Aging 4, 60–72. doi:10.1016/j.tma.2020.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Varma, H., Cheng, R., Voisine, C., Hart, A. C., and Stockwell, B. R. (2007). Inhibitors of Metabolism rescue Cell Death in Huntington's Disease Models. Proc. Natl. Acad. Sci. U S A. 104, 14525–14530. doi:10.1073/pnas.0704482104

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaughn, A. R., Haas, K. N., Burney, W., Andersen, E., Clark, A. K., Crawford, R., et al. (2017). Potential Role of Curcumin against Biofilm-Producing Organisms on the Skin: A Review. Phytother Res. 31, 1807–1816. doi:10.1002/ptr.5912

PubMed Abstract | CrossRef Full Text | Google Scholar

Ved, R., Saha, S., Westlund, B., Perier, C., Burnam, L., Sluder, A., et al. (2005). Similar Patterns of Mitochondrial Vulnerability and rescue Induced by Genetic Modification of Alpha-Synuclein, Parkin, and DJ-1 in Caenorhabditis elegans. J. Biol. Chem. 280, 42655–42668. doi:10.1074/jbc.M505910200

PubMed Abstract | CrossRef Full Text | Google Scholar

Verma, R., Gurumurthy, M., Yeo, B. C. M., Lu, Q., Naftalin, C. M., and Paton, N. I. (2022). Effects of Increasing Concentrations of Rifampicin on Different Mycobacterium tuberculosis Lineages in a Whole-Blood Bactericidal Activity Assay. Antimicrob. Agents Chemother. 66, AAC0169921. doi:10.1128/AAC.01699-21

CrossRef Full Text | Google Scholar

Vieira, F. G., Ping, Q., Moreno, A. J., Kidd, J. D., Thompson, K., Jiang, B., et al. (2015). Guanabenz Treatment Accelerates Disease in a Mutant SOD1 Mouse Model of ALS. Plos One 10, e0135570. doi:10.1371/journal.pone.0135570

PubMed Abstract | CrossRef Full Text | Google Scholar

Viszwapriya, D., Subramenium, G. A., Prithika, U., Balamurugan, K., and Pandian, S. K. (2016). Betulin Inhibits Virulence and Biofilm of Streptococcus Pyogenes by Suppressing ropB Core Regulon, sagA and dltA. Pathog. Dis. 74. doi:10.1093/femspd/ftw088

PubMed Abstract | CrossRef Full Text | Google Scholar

Voisine, C., Varma, H., Walker, N., Bates, E. A., Stockwell, B. R., and Hart, A. C. (2007). Identification of Potential Therapeutic Drugs for huntington's Disease Using Caenorhabditis elegans. Plos One 2, e504. doi:10.1371/journal.pone.0000504

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Saar, V., Leung, K. L., Chen, L., and Wong, G. (2018). Human Amyloid β Peptide and Tau Co-expression Impairs Behavior and Causes Specific Gene Expression Changes in Caenorhabditis elegans. Neurobiol. Dis. 109, 88–101. doi:10.1016/j.nbd.2017.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Lau, C. Y., Ma, F., and Zheng, C. (2021a). Genome-wide Screen Identifies Curli Amyloid Fibril as a Bacterial Component Promoting Host Neurodegeneration. Proc. Natl. Acad. Sci. U.S.A. 118. doi:10.1073/pnas.2106504118

CrossRef Full Text | Google Scholar

Wang, H., Liu, X., Tan, C., Zhou, W., Jiang, J., Peng, W., et al. (2020a). Bacterial, Viral, and Fungal Infection-Related Risk of Parkinson's Disease: Meta-Analysis of Cohort and Case-Control Studies. Brain Behav. 10, e01549. doi:10.1002/brb3.1549

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, I. F., Guo, B. S., Liu, Y. C., Wu, C. C., Yang, C. H., Tsai, K. J., et al. (2012). Autophagy Activators rescue and Alleviate Pathogenesis of a Mouse Model with Proteinopathies of the TAR DNA-Binding Protein 43. Proc. Natl. Acad. Sci. U S A. 109, 15024–15029. doi:10.1073/pnas.1206362109

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Farr, G. W., Hall, D. H., Li, F., Furtak, K., Dreier, L., et al. (2009). An ALS-Linked Mutant SOD1 Produces a Locomotor Defect Associated with Aggregation and Synaptic Dysfunction when Expressed in Neurons of Caenorhabditis elegans. Plos Genet. 5, e1000350. doi:10.1371/journal.pgen.1000350

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, N., Zhou, Y., Zhao, L., Wang, C., Ma, W., Ge, G., et al. (2020b). Ferulic Acid Delayed Amyloid β-induced Pathological Symptoms by Autophagy Pathway via a Fasting-like Effect in Caenorhabditis elegans. Food Chem. Toxicol. 146, 111808. doi:10.1016/j.fct.2020.111808

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, N. Y., Li, J. N., Liu, W. L., Huang, Q., Li, W. X., Tan, Y. H., et al. (2021b). Ferulic Acid Ameliorates Alzheimer's Disease-like Pathology and Repairs Cognitive Decline by Preventing Capillary Hypofunction in APP/PS1 Mice. Neurotherapeutics 18, 1064–1080. doi:10.1007/s13311-021-01024-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Watson, E., MacNeil, L. T., Ritter, A. D., Yilmaz, L. S., Rosebrock, A. P., Caudy, A. A., et al. (2014). Interspecies Systems Biology Uncovers Metabolites Affecting C. elegans Gene Expression and Life History Traits. Cell 156, 1336–1337. doi:10.1016/j.cell.2014.02.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Williamson, T. P., Johnson, D. A., and Johnson, J. A. (2012). Activation of the Nrf2-ARE Pathway by siRNA Knockdown of Keap1 Reduces Oxidative Stress and Provides Partial protection from MPTP-Mediated Neurotoxicity. Neurotoxicology 33, 272–279. doi:10.1016/j.neuro.2012.01.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Wong, S. Q., Pontifex, M. G., Phelan, M. M., Pidathala, C., Kraemer, B. C., Barclay, J. W., et al. (2018). α-Methyl-α-phenylsuccinimide Ameliorates Neurodegeneration in a C. elegans Model of TDP-43 Proteinopathy. Neurobiol. Dis. 118, 40–54. doi:10.1016/j.nbd.2018.06.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, J., Xu, H., Tang, W., Kopelman, R., Philbert, M. A., and Xi, C. (2009). Eradication of Bacteria in Suspension and Biofilms Using Methylene Blue-Loaded Dynamic Nanoplatforms. Antimicrob. Agents Chemother. 53, 3042–3048. doi:10.1128/AAC.01604-08

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Park, K. C., Choi, B. G., Park, J. H., and Yoon, K. S. (2016). The Antibiofilm Effect of Ginkgo Biloba Extract against Salmonella and Listeria Isolates from Poultry. Foodborne Pathog. Dis. 13, 229–238. doi:10.1089/fpd.2015.2072

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Wu, Z., Butko, P., Christen, Y., Lambert, M. P., Klein, W. L., et al. (2006). Amyloid-beta-induced Pathological Behaviors Are Suppressed by Ginkgo Biloba Extract EGb 761 and Ginkgolides in Transgenic Caenorhabditis elegans. J. Neurosci. 26, 13102–13113. doi:10.1523/JNEUROSCI.3448-06.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, L., Li, H., Zhang, J., Yang, F., Huang, A., Deng, J., et al. (2014). Salidroside Protects Caenorhabditis elegans Neurons from Polyglutamine-Mediated Toxicity by Reducing Oxidative Stress. Molecules 19, 7757–7769. doi:10.3390/molecules19067757

PubMed Abstract | CrossRef Full Text | Google Scholar

Xin, L., Yamujala, R., Wang, Y., Wang, H., Wu, W. H., Lawton, M. A., et al. (2013). Acetylcholineestarase-inhibiting Alkaloids from Lycoris Radiata Delay Paralysis of Amyloid Beta-Expressing Transgenic C. elegans CL4176. Plos One 8, e63874. doi:10.1371/journal.pone.0063874

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamada, K. J., Heim, C. E., Xi, X., Attri, K. S., Wang, D., Zhang, W., et al. (2020). Monocyte Metabolic Reprogramming Promotes Pro-inflammatory Activity and Staphylococcus aureus Biofilm Clearance. Plos Pathog. 16, e1008354. doi:10.1371/journal.ppat.1008354

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, X., Zhang, M., Dai, Y., Sun, Y., Aman, Y., Xu, Y., et al. (2020). Spermidine Inhibits Neurodegeneration and Delays Aging via the PINK1-PDR1-dependent Mitophagy Pathway in C. elegans. Aging (Albany NY) 12, 16852–16866. doi:10.18632/aging.103578

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, C., Johnson, W. M., Gao, Y., Wang, W., Zhang, J., Deak, M., et al. (2013). Kinase Inhibitors Arrest Neurodegeneration in Cell and C. elegans Models of LRRK2 Toxicity. Hum. Mol. Genet. 22, 328–344. doi:10.1093/hmg/dds431

PubMed Abstract | CrossRef Full Text | Google Scholar

You, Z., Ran, X., Dai, Y., and Ran, Y. (2018). Clioquinol, an Alternative Antimicrobial Agent against Common Pathogenic Microbe. J. Mycol. Med. 28, 492–501. doi:10.1016/j.mycmed.2018.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

You, Z., Zhang, C., and Ran, Y. (2020). The Effects of Clioquinol in Morphogenesis, Cell Membrane and Ion Homeostasis in Candida Albicans. BMC Microbiol. 20, 165. doi:10.1186/s12866-020-01850-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaidi, S., Singh, S. L., and Khan, A. U. (2020). Exploring Antibiofilm Potential of Bacitracin against streptococcus Mutans. Microb. Pathog. 149, 104279. doi:10.1016/j.micpath.2020.104279

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, C., Liu, Z., Bunker, E., Ramirez, A., Lee, S., Peng, Y., et al. (2017). Sorafenib Targets the Mitochondrial Electron Transport Chain Complexes and ATP Synthase to Activate the PINK1-Parkin Pathway and Modulate Cellular Drug Response. J. Biol. Chem. 292, 15105–15120. doi:10.1074/jbc.M117.783175

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, D., Anantharam, V., Kanthasamy, A., and Kanthasamy, A. G. (2007). Neuroprotective Effect of Protein Kinase C delta Inhibitor Rottlerin in Cell Culture and Animal Models of Parkinson's Disease. J. Pharmacol. Exp. Ther. 322, 913–922. doi:10.1124/jpet.107.124669

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Su, Y., Sun, Z., Chen, M., Han, Y., Li, Y., et al. (2021). Ginsenoside Rg1 Alleviates Aβ Deposition by Inhibiting NADPH Oxidase 2 Activation in APP/PS1 Mice. J. Ginseng Res. 45, 665–675. doi:10.1016/j.jgr.2021.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S. Q., Obregon, D., Ehrhart, J., Deng, J., Tian, J., Hou, H., et al. (2013). Baicalein Reduces β-amyloid and Promotes Nonamyloidogenic Amyloid Precursor Protein Processing in an Alzheimer's Disease Transgenic Mouse Model. J. Neurosci. Res. 91, 1239–1246. doi:10.1002/jnr.23244

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W., He, H., Song, H., Zhao, J., Li, T., Wu, L., et al. (20162016). Neuroprotective Effects of Salidroside in the MPTP Mouse Model of Parkinson's Disease: Involvement of the PI3K/Akt/GSK3β Pathway. Parkinsons Dis. 2016, 9450137. doi:10.1155/2016/9450137

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, W. X., Zhang, J. H., Cao, J. B., Wang, W., Wang, D. X., Zhang, X. Y., et al. (2017). Acetaminophen Attenuates Lipopolysaccharide-Induced Cognitive Impairment through Antioxidant Activity. J. Neuroinflammation 14, 17. doi:10.1186/s12974-016-0781-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, C., Karimzadegan, S., Chiang, V., and Chalfie, M. (2013). Histone Methylation Restrains the Expression of Subtype-specific Genes during Terminal Neuronal Differentiation in Caenorhabditis elegans. Plos Genet. 9, e1004017. doi:10.1371/journal.pgen.1004017

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, T., Zhu, M., and Liang, Z. (2018). (-)-Epigallocatechin-3-gallate Modulates Peripheral Immunity in the MPTP-Induced Mouse Model of Parkinson's Disease. Mol. Med. Rep. 17, 4883–4888. doi:10.3892/mmr.2018.8470

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Y., Smith, D., Leong, B. J., Brännström, K., Almqvist, F., and Chapman, M. R. (2012). Promiscuous Cross-Seeding between Bacterial Amyloids Promotes Interspecies Biofilms. J. Biol. Chem. 287, 35092–35103. doi:10.1074/jbc.M112.383737

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: neurodegenerative diseases, Caenorhabditis elegans, gut microbiome, curli fibers biofilm, csgA gene, Parkinson’s disease, disease modeling, microbe-host interaction

Citation: Wang C and Zheng C (2022) Using Caenorhabditis elegans to Model Therapeutic Interventions of Neurodegenerative Diseases Targeting Microbe-Host Interactions. Front. Pharmacol. 13:875349. doi: 10.3389/fphar.2022.875349

Received: 14 February 2022; Accepted: 08 March 2022;
Published: 28 April 2022.

Edited by:

Long Ma, Central South University, China

Reviewed by:

Matthew Richard Chapman, University of Michigan, United States
Christopher D. Link, University of Colorado Boulder, United States

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

*Correspondence: Chaogu Zheng, cgzheng@hku.hk

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