Next Article in Journal
Impact of Growth Conditions on the Viability of Trichoderma asperellum during Storage
Next Article in Special Issue
Antifungal Activity of Cell-Free Filtrate of Probiotic Bacteria Lactobacillus rhamnosus ATCC-7469 against Fungal Strains Isolated from a Historical Manuscript
Previous Article in Journal
Identification of Fusarium oxysporum f. sp. lactucae Race 1 as the Causal Agent of Lettuce Fusarium Wilt in Greece, Commercial Cultivars’ Susceptibility, and Temporal Expression of Defense-Related Genes
Previous Article in Special Issue
The Preventive Effects of Probiotic Prevotella histicola on the Bone Loss of Mice with Ovariectomy-Mediated Osteoporosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics for Neurodegenerative Diseases: A Systemic Review

1
Cell & Developmental Biology Laboratory, Centre of Research for Development, Parul University, Vadodara 391760, India
2
Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
3
The Neotia University, Sarisha 743368, India
4
Instituto de Conservacióny Mejora de la Agrodiversidad Valenciana, Universitat Politècnica de València, 46022 Valencia, Spain
*
Authors to whom correspondence should be addressed.
Microorganisms 2023, 11(4), 1083; https://doi.org/10.3390/microorganisms11041083
Submission received: 15 March 2023 / Revised: 12 April 2023 / Accepted: 18 April 2023 / Published: 20 April 2023
(This article belongs to the Special Issue Effects of Probiotics on Health)

Abstract

:
Neurodegenerative disorders (ND) are a group of conditions that affect the neurons in the brain and spinal cord, leading to their degeneration and eventually causing the loss of function in the affected areas. These disorders can be caused by a range of factors, including genetics, environmental factors, and lifestyle choices. Major pathological signs of these diseases are protein misfolding, proteosomal dysfunction, aggregation, inadequate degradation, oxidative stress, free radical formation, mitochondrial dysfunctions, impaired bioenergetics, DNA damage, fragmentation of Golgi apparatus neurons, disruption of axonal transport, dysfunction of neurotrophins (NTFs), neuroinflammatory or neuroimmune processes, and neurohumoral symptoms. According to recent studies, defects or imbalances in gut microbiota can directly lead to neurological disorders through the gut-brain axis. Probiotics in ND are recommended to prevent cognitive dysfunction, which is a major symptom of these diseases. Many in vivo and clinical trials have revealed that probiotics (Lactobacillus acidophilus, Bifidobacterium bifidum, and Lactobacillus casei, etc.) are effective candidates against the progression of ND. It has been proven that the inflammatory process and oxidative stress can be modulated by modifying the gut microbiota with the help of probiotics. As a result, this study provides an overview of the available data, bacterial variety, gut-brain axis defects, and probiotics’ mode of action in averting ND. A literature search on particular sites, including PubMed, Nature, and Springer Link, has identified articles that might be pertinent to this subject. The search contains the following few groups of terms: (1) Neurodegenerative disorders and Probiotics OR (2) Probiotics and Neurodegenerative disorders. The outcomes of this study aid in elucidating the relationship between the effects of probiotics on different neurodegenerative disorders. This systematic review will assist in discovering new treatments in the future, as probiotics are generally safe and cause mild side effects in some cases in the human body.

1. Introduction

Neurodegenerative diseases are a group of disorders characterized by the progressive loss of structure and function of neurons in the central and/or peripheral nervous system [1]. These diseases affect various parts of the brain, spinal cord, and nerves, leading to a gradual decline in cognitive, motor, and/or sensory function [2]. There is emerging evidence to suggest that probiotics, which are live microorganisms that provide health benefits when consumed in adequate amounts, may have a role to play in the prevention and treatment of neurodegenerative diseases [3]. Studies regarding probiotics suggest that utilization of probiotics increased intestinal flora which can regulate the inflammatory response, as well as resist the colonization of exogenous pathogenic microorganisms [4].
While probiotics are generally considered safe and can provide numerous health benefits, there are several limitations to their use. Here are some of the most important ones:
(1) Limited evidence for specific health conditions: While probiotics have been shown to be effective in treating certain conditions, such as diarrhea caused by antibiotics, there is limited evidence to support their use in treating other health conditions. For example, while some studies have suggested that probiotics may be beneficial for conditions such as irritable bowel syndrome (IBS) and eczema, the evidence is not yet strong enough to make definitive recommendations [5]. (2) Variability in strains and dosages: Different strains of probiotics can have different effects on the body, and the appropriate dosage can vary depending on the specific strain being used. Therefore, it can be difficult to determine the optimal strain and dosage for a particular health condition, and the effectiveness of probiotics can vary widely depending on these factors [6]. (3) Limited survival and colonization: Probiotic bacteria must survive the acidic environment of the stomach and colonize the gut in order to provide health benefits. However, not all probiotic strains are able to survive this process, and even those that do may have difficulty colonizing the gut in sufficient numbers to have a meaningful effect [7]. (4) Short-term effects: Probiotics are typically used for short periods of time, often a few weeks or months. However, the long-term effects of probiotic use are not well understood, and it is possible that their benefits may diminish over time [7]. (5) Potential adverse effects: While probiotics are generally considered safe, they can sometimes cause adverse effects such as bloating, gas, and diarrhea. In rare cases, they may even cause serious infections, particularly in people with weakened immune systems [8]. However, contrary to its side effects, it is highly effective, and healthy gut microbiota can help reduce inflammation and oxidative stress, which are two key factors that contribute to the development of neurodegenerative diseases [9]. Additionally, some probiotics have been shown to create molecules that can stimulate the production of brain-derived neurotrophic factor (BDNF), a protein that promotes the growth and survival of neurons in the brain. Low levels of BDNF have been linked to several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease [10]. While the research on probiotics and neurodegenerative diseases is still in its early stages, there is growing interest in exploring the potential benefits of probiotics in this area. However, more research is needed to determine the optimal strains, dosages, and treatment regimens for different neurodegenerative diseases.
A systematic review is a type of research study that collects and analyzes all available evidence on a specific topic, in order to answer a specific research question [11]. In the case of probiotics and neurodegenerative diseases, a systematic review can help to provide a comprehensive overview of the current state of research on this topic. There are several reasons why a systematic review is important for examining the effects of probiotics on neurodegenerative diseases. Firstly, there is a large and growing body of research on this topic, and a systematic review can help to synthesize this information and identify the most promising areas for future research. Secondly, neurodegenerative diseases are complex and multifactorial disorders, and it is likely that probiotics may have different effects on different aspects of these diseases. For example, probiotics may have different effects on cognitive function, motor symptoms, and inflammation in different neurodegenerative diseases [12]. A systematic review can help to identify these differences and provide guidance on the most appropriate use of probiotics for different patient populations. Thirdly, there is currently a lack of consensus on the optimal probiotic strains and dosages for different neurodegenerative diseases. A systematic review can help to identify the most effective probiotic interventions and inform the development of future clinical trials. In summary, a systematic review is an important tool for evaluating the effects of probiotics on neurodegenerative diseases, as it can provide a comprehensive and evidence-based overview of the current state of research on this topic.

2. Methods

2.1. Literature Search Strategy

The recommended notification items for systematic reviews and meta-analyses (PRISMA) recommendations are used to direct the literature search [13]. Three electronic databases were searched from inception to March 2023: MEDLINE/PubMed, Springer Link, and Nature with Probiotics and Neurodegenerative diseases keywords. No publication year restrictions were applied and only English literatures and Free open access literature were used.

2.2. Inclusion and Exclusion Criteria

Inclusion and Exclusion criteria used in this systematic study is described in below Table 1.

2.3. Management of Extracted Data

Literature screening is based on Figure 1. Each article’s abstract and title are first separately reviewed by two evaluators. (N.P. and S.O.). Each of the papers that were chosen had to have covered one of the following metrics that were mentioned in the selection criteria. Any discrepancy is resolved with discussion and consultation of third reviewer (M.J. and P.K.). No replication or dispersion data are mentioned for in vivo experiments. In vivo studies are found to be free of bias, in contrast to clinical trials, which are subject to a bias evaluation. We assure that there is no duplication of any literature.

2.4. Strategy of Data Extraction

The data extraction provides the results of the chosen papers describe in tabular format in Section 4.6, while the study’s findings were discussed in Section 3. The Section 4 explained how the results were analyzed.

2.5. Data Synthesis and Statistical Analysis

If there was heterogeneity between articles, data were combined using a random effects model to provide a more cautious estimate of the spectrum of probiotics in neurodegenerative diseases [14]. Both the I2 index 50% and the chi-squared test with a p < 0.05, used to identify a significant degree of heterogeneity, were used to evaluate heterogeneity, which is variance between individual research findings that has not occurred by accident [15]. All statistical analysis was conducted through MS Excel (Windows XPC) by reviewer M.J.

3. Results

3.1. Literature Search and Selection

An initial search of the literature identified 2264 potentially related articles. Due to the free access of articles, 1521 were excluded from the study. The remaining 743 articles were further evaluated on the basis of duplication and 86 articles were removed. Around 487 articles are removed as they are under the exclusion criteria of Case reports, Hypothesis, Communication, Systematic review and Current opinion. The final 52 articles are full texts and eligible for the systematic review and discussion.

3.2. Characteristics of the Included Studies

We identified 20 articles regarding Parkinson’s disease and 32 Articles regarding Alzheimer’s disease with probiotics that have reported the potential effect of probiotics in neurodegenerative diseases. Table 2 describes the selected studies according to the type of probiotics, their effects, and their mechanism of action in particular neurodegenerative diseases.

4. Discussion

4.1. Gut Microbiota-Brain Axis

The gut-brain axis is a complicated biochemical pathway that connects the gastrointestinal tract and the brain, it is a bi-directional interaction between GI and CNS microbiota via endocrine, neural, humoral, and immune links Figure 2 [16].
This communication is responsible for both healthy and unhealthy benefits. The diversity of gut microbiota has both pathogenic and commensal roles, such as Lactobacillus releasing short-chain fatty acids and acetylcholine. Whereas Bacillus produced norepinephrine and dopamine [17]. The gut microbiota performed some functions repeatedly in a cyclic way in the brain such as regulation of the hypothalamic-pituitary-adrenal axis, which released cortisol that is going to activate the brain microglia and releases cytokines. They also control the PNS and CNS by establishing a connection between vagal nerves, adrenergic nerve activation, and several immunomodulatory, neuropeptides, endocrine hormones, and neurotransmitters. Gut microbiota diversity and their action can be affected by hormones such as noradrenaline [18]. If the gut epithelial barrier is being dysregulated, it disturbed the brain-gut-microbiota axis, which encourages the production of neuroactive substances and neurotropic viruses that regulates pathogens with slow neurotoxic properties [19]. The immune responses of the gut are harmonized by the microbial community present in the gut by various cells that maintain various immunological conditions. Microbiota of the gut can be analyzed by Next-generation sequencing and meta-genome analysis reveals that the mammalian gut has microbiomes (most commonly Cryptosporidium sp. Shigella sp., Enterotoxigenic Escherichia coli) [20]. Bacteria are hardly able to pass the blood-brain-barrier or blood-cerebrospinal fluid barrier to enter CNS, by the mechanism called trans-cellular infiltration, infected leukocytes, and para-cellular entering [21]. The mechanism that may influence neurodegeneration through gut microbiota, is the production of functional metabolites transferred through vagus nerves, microbial-associated molecular patterns, and unable to inhibit harmful gut microbes. The pathological process of the gut microbiome is to release neurotoxic metabolites that can be transferred through the gut-brain axis or microbial-molecular patterns, and are unstoppable by the peripheral immune system [22].

4.2. Relation of Gut Microbiota with Neurodegenerative Disorders

The health of the brain is synchronized or regulated by the GI tract or it is directly proportional to the microbiome present in the human gut. Imbalance in the microbial community is associated with many diseases, but in the brain, it was responsible for neurodegenerative diseases such as AD, PD, and others disorders [23]. Brain and gut microbiota can interrelate with each other through several pathways such as neuroendocrine, neuroimmune, and autonomic nervous systems. Interactive partners that perform the mechanisms are the cell wall, neurotransmitters, vagus nerves, and metabolites [24]. The microbiome of the gut can synthesize the neurotransmitter that may help to maintain the homeostasis of the central nervous system, which can influence neurodegeneration, examples are tryptophan, brain-derived neurotrophic factor (BDNF), Gama-aminobutyric acid (GABA), and short-chain fatty acid (SCFA) [25]. Neurotrophins have a neuroprotective property that is important for the growth, development, and synaptic plasticity along with the differentiation and survival of the neuron. The decreased level of BDNF influences the neurodegenerative disease related to the cerebral cortex and directly relates to the gut-brain axis, which triggers other diseases too [26]. Gut microbes such as Faecalibacterium prausnitzil, Clostridium leptum and Eubacterium rectala, etc. produce short-chain fatty acids through a down regulation of pro-inflammatory cytokines that play a major role in neurodegeneration. Microbial-derived SCFAs are produced by bacterial fermentation and have neuro-active functions, they act as a modulator for serotonin (neurotransmitter) and some neuropeptides which help to facilitate the gut-brain axis at various stages. During the release mechanism which influences neuronal health and behavioral response [27]. Excessive release of SCFAs is responsible for neuronal health and behavioral responses. An essential amino acid called tryptophan plays an important role in the synthesis of serotonin and other neurotransmitters in the CNS. Imbalance in their levels leads to brain and gastrointestinal disturbances that may cause neurodegeneration, cognitive impairment, and mood disorders [28,29]. An important inducer that administered the excitation of the neurons, is a by-product of bacterial metabolism called GABA. Deregulation of GABA leads to various pathological imbalances that play a major role in neuro-cytotoxicity which accelerates several chronic neurological disorders. GABA is an example that proves how gut microbiota regulates brain chemistry [30].

4.2.1. Alzheimer’s Disease (AD)

People with gut disorders more prone to have AD in the future. Changes in complex ecosystems are co-related with many gastrointestinal disturbances that can implicate many inflammatory diseases including obesity, diabetes and inflammatory bowel disease. The dysbiosis of gut microbiota has an impact on the synthesis of proteins and metabolic processes which are related to the progression of the disease such as AD. Aging changes the gut microbial concentration which enhances pro-inflammatory bacterial growth more than anti-inflammatory bacteria that deteriorates the permeability of the blood-brain barrier and GIT (Gastro Intestinal Tract) functions [31]. Pro-inflammatory phylum such as Proteobacteria, Verrucomicrobia, genera such as Escherichia/Shigella, and species such as Pseudomonas aeruginosa, anti-inflammatory species are Clostridium spp., and Ruminococcaceae. Some study reveals that increased mRNA encoding initiates the release of pro-inflammatory cytokines such as, IL6, CXCL2, and NLRP3, Escherichia/Shigella are related to pro-inflammatory taxon [28]. The presence of Helicobacter pylori in the gut microbiota increases the release of inflammatory mediators which increases the amyloid β 40/42 ratio in the serum, other bacteria such as Borrelia burgdorferi and Chlamydia pneumoniae also participate in the hyper phosphorylation of tau which is an important hallmark of AD. IL-10 is an anti-inflammatory cytokine Eubacterium rectale is associated with an anti-inflammatory taxon [32].

4.2.2. Parkinson’s Disease

PD is a multifocal neuronal disease that is distinguished by tremors, slow movement, akinesia, muscular rigidity, gait, and difficulty in walking. Instead of these symptoms, PD patients suffer from constipation which is one of the causes of increased intestinal permeability and inflammation that is directly related to the microbiota community of the small intestine [33]. Small intestinal bacterial outgrowth and helicobacter pylori infection has seen in diseased person that causes motor impairment and problem-related with stool [34]. In most cases, patients suffer from increased mucosal permeability and endo-toxic exposure caused by Coliform bacteria [35]. Comparably, some bacteria are reduced in feces such as Roseburia intestinalis, Roseburia hominis, Coprococcus eutactus, and Blautia faecis dysregulate the biosynthesis of lipopolysaccharides and are also responsible for GABA deregulation [36]. Escherichia coli is a Gram-negative bacteria that releases amyloidogenic protein which induces alpha-synuclein aggregation and which regulates disease in the gut and neurodegeneration in the brain [37].

4.2.3. Huntington’s Disease

HD is a genetic disease caused by overexpression of the huntingtin coding gene, new research suggests that an imbalance in gut microbiota dysregulates the cytokine levels and excessive production of hydrogen sulfide that negatively affects gut health [38,39]. Imbalance is seen in two microbial communities such as increased the majority of the bacterial phyla Bacteroidetes (4%) and Firmicutes (83%) which causes mortality and motor ability-related problems in HD conditions. Research suggests that irregular intestinal biomicrome decreases mucosal thickness and decreased neuropeptide formation with abnormal endocrine hormonal conditions [40,41,42]. ATP levels are associated with Prevotella scopos which has negative effects and also has a co-relation with decreased butyrate formation affected by Blautia producta [43].

4.2.4. Other Neurological Disorders

In neurological disorders, the nerve function and structure become affected badly, causes of neuronal cell death, are Amyotrophic lateral sclerosis, Friedreich ataxia, Lewy body disease, spinal muscular atrophy, and Epilepsy, etc. Epilepsy, a neuro-psychiatric disorder is a result of environmental and genetic imbalance, several studies implicated dysbiosis of gut microbiota correlated with the disease, and imbalance of microbiome increases the pro, and anti-inflammatory effects, which leads to chronic inflammation and progression of the disease [44,45]. Bacteria out-growth downregulates lipid and glucose metabolism which disturbs the ATP binding cassette and transporter-associated pathways [46]. Amyotrophic lateral sclerosis is a degenerative condition that is caused by the mutation in dozens of genes which produces a misfolded protein that is found in motor neurons, responsible for voluntary muscular movement [47]. Bulbar function slowly deteriorated with the progression of the disease, and dysphagia (because of aspiration pneumonia and weight loss) have seen [48]. ALS is implicated by the deregulation of the resident and peripheral immune system. Gut microbiota connected with the intestinal immune system, because of invasion or dysbiotic leaky gut and disturbed molecular patterns, provoke cells to release pro-inflammatory cytokines that deregulate the Firmicutes/Bacteroidetes ratio [49]. Bacteroidetes are good gut bacteria, that are decreasing at the diseased condition that imbalances the cell homeostasis, is regarded as dysbiosis and several reviews suggest that the gut plays an impotent role in the progression of Lewy body disease, and intestinal pro-inflammation is the cause of the chronic phase of the disease. An imbalance in microbiota and translocation of the lumen through a leaky gut are key mechanisms of the disease [50].

4.3. Elucidation the Role of Gut Microbiota in Neuroinflammation

Activation of immune cells defined as a pro-inflammatory condition, is an important pathophysiological aspect, behind all neurodegenerative disorders, and leads to neuroinflammation, mediated by the secretion of chemokine, cytokine, ROS, and secondary messengers [51]. CNS is protected from all kinds of toxins through the blood-brain barrier, and its disruptive permeability accumulates neurotoxin waste that increases immune cell influxes [52]. Colonization of Helicobacter pylori is responsible for dysbiosis in the gut and causes systemic inflammation that accelerates pathologies for neurodegeneration and other disorder such as asthma, metabolic syndrome, and allergy. The phyla that constitute this core microbiota are Bacteroides and Firmicutes, which are the dominant phylum, in addition to Lentisphaerota, Actinomycetota, Spirochaetota, Verrucomicrobiota, Proteobacteria, and Fusobacteriota. Microbiomes produce metabolites involved in cell homeostasis such as SCFAs regulate the homeostasis of microglia, and induced T cell activation to fight autoimmunity [53]. Tryptophan metabolism facilitates the CNS-microbiome interaction through metabolic by-products such as indole tryptamine, indole acetic acid, quinolinate, and indole propionic acid are neuroactive metabolites and able to regulate CNS activity [54]. These metabolites enter in CNS through the leaky gut epithelium and alteration in Blood-Brain-Barrier facilitates the endotoxins to enter into CNS. Vegas nerves, play an important role to define CNS-microbiome relations, are directly connected with the gut and brain, and are also able to sense bacterial metabolites by their afferent composition. Vegas nerve transfers this information, which has concerns about altered microbiota to CNS, so that appropriate action should be initiated to avoid neuropathological conditions [55]. Blood-brain-barrier designed to protect CNS from the toxin and pathogens that can damage the parenchymal lining and disrupts its working which allows the entry of neurotoxins and microbial metabolic wastes into the CNS, which can be one of the causes of neuroinflammation [56].

4.4. Understanding the Role of Probiotics in Gut Microbiota

Probiotics were first proposed by Nobel Prize recipient Elie Metchnikoff at the turn of the 20th century. According to today’s meaning, probiotics are living microorganisms that, when given to a host in sufficient quantities, help their health Figure 3 [57]. The management of gut microbial communities, the repression of pathogens, immunomodulation, promotion of epithelial cell proliferation and differentiation, and reinforcement of the digestive barrier are some of the mechanisms of probiosis [58]. The connections between gut microbes and the immune system have revealed previously unknown microbial components or receptors that, in addition to the conventional immune components, also regulate energy, glucose, and lipid metabolism, in reference to that Table 2.
Cani et al., 2007 introduced the idea of metabolic endotoxemia. Notably, a slight increase in blood LPS was found to be a crucial factor in the onset of low-grade inflammation, and eventually insulin resistance in models of genetically predisposed or diet-induced obesity and diabetes, as well as related cardio metabolic diseases [59]. The relevant and significant literature concentrates mainly on different strains, from more recent prospects such as A. muciniphila and Faecalibacterium prausnitzii are considered to be superior to more traditional probiotics such as Lactobacillus and Bifidobacterium or the yeast Saccharomyces boulardii [60]. According to research by Bo et al., 2020, Bifidobacterium pseudolongum was able to reverse the dysbiosis of the gut microbiota in obese mice, including the variety of the microbiome and the proportion of Firmicutes to Bacteroidetes [61]. The bacterial species Bifidobacterium and Butyricimonas were also more prevalent after this therapy. Furthermore, the defense system has an impact on the central nervous system (CNS). In light of this, immunity has a direct impact on people’s quality of life, and controlling gut bacterial populations with probiotics has proven to be a successful strategy for doing so [62]. This systematic study reviews the impact of probiotics on human immunity and the gut microbiome in relation to neurodegenerative diseases (mainly AD, PD, and HD).
Table 2. Popular probiotic strains, their location in the gut, function in the human body, and health benefits with their source (food).
Table 2. Popular probiotic strains, their location in the gut, function in the human body, and health benefits with their source (food).
Strain NameLocation in GutFunctionSource (Food)Health
Benefits
Refs.
Bifidobacterium bifidumInterior intestineActivate the host immunity, adhere to gut mucosa, and metabolize host glycan (mucin)Yogurt, Kefir, Sauerkraut, GarlicPrevent inflammation, Enteric cancer, Ulcerative colitis and depression[63,64]
Bifidobacterium breveGastrointestinal tract Modulating expression of inflammatory receptorsKombucha, water kefir, and raw sauerkrautPrevent pediatrics, For pathologies such as diarrhea and infant colics, to celiac disease, obesity, allergic and neurological disorders[65,66,67]
Bifidobacterium longumInterior intestineInhibits inflammation by regulating the immune system, improving the intestinal barrier function, and increasing acetate productionGoat dairy products, such as yogurt, kefir, seaweed, and miso soupReduced stress and improved memory, improving irritable bowel syndrome[68,69,70]
Bifidobacterium animalisMajorly in animal intestineReduce the inflammatory receptor expressionMammalian colon and MilkConstipation, irritable bowel syndrome (IBS), respiratory system infections, and excessive screaming in infants[71,72]
Bifidobacterium catenulatumOn the wall of GIMaintain functional integrity of gutMostly found in breast-fed infantsFolate production in the intestines of infants mainly[73,74]
Bifidobacterium pseudocatenulatumStomachIntestine cancer prevention, Enhancement of host immune responses, maintain liver functionalityMilk, dairy products, and other carbohydrate source such as xylan or arabinoxylanModulate the gut–bone axis, inhibit inflammation, blocking Pro inflammatory Cytokines, Inhibiting TLR4/NF-κB Signaling[75,76,77]
Akkermansia muciniphilaResides in the mucus layer of the large intestineIncreasing mucus thickness and increasing gut barrier functionCranberries, grapes, black tea, and walnutsProtecting and strengthening your gut lining, prevents inflammation, manage glucose level in body[78,79,80]
Faecalibacterium prausnitziiInside the intestineHigh production of SCFAs that escape digestion and absorption in small intestineFruits and vegetables such as chicory roots, wheat, onion, banana, garlic, and leekWeakened intestinal anti-inflammatory and immune regulation capabilities.[81]
Lactobacillus acidophilusStomach, duodenum, and jejunum Inhibiting carcinogen and mutagen formation, altering overall metabolismMilk enriched with acidophilus, yoghurt, miso, and tempehTreat bacterial vaginosis, yeast infection, digestive disorder, and some neurological disorder[82,83]
Lactococcus lactisPassage of GIImproved the growth performance and regulated amino acid profiles, intestinal immunity, and microbiota in weaning pigletsYogurt, cheese, and sauerkrautExhibit protection against non-respiratory pathogens, such as HIV, Human papillomavirus and the malarial parasite[84]
Lactiplantibacillus plantarumSmall intestine barrierImmunomodulating properties and decrease the anti-inflammatory cytokineKimchi, Ogi, sourdough, and fermented plant material, and fermented sausagesAntioxidant, cancer-preventative, anti-inflammatory, antiproliferative, anti-obesity, and anti-diabetic properties[85]
Clostridium butyricumIntestinal tractIntestinal microbiota disorder in human and enhance the humoral immune responseSoured milk and cheeses.suppress inflammatory cytokine secretion, and modulate CNS autoimmunity, inhibit the increase in IL-17A gene expression[86,87]
E. coliLower intestineKeep digestive system healthy, breakdown and digestion of foodRaw vegetables and undercooked ground beef and contaminated waterGrowth of tumor inhibit by ClyA toxin (E. coli), decreased the postprandial blood glucose, antipyretic, anti-inflammatory, and anti-amyloidic[88]

4.5. Recent Evidence for Probiotics and Neurodegenerative Disorders

In neurodegenerative disorders, neuroinflammation plays a very crucial role in pathogenesis, which has been proven through various studies. That may further drive the progressive loss of dopaminergic neurons. Therefore, increasing efforts on anti-inflammation approaches are being made in developing a cure for ND [89]. Different probiotics such as E. coli, Lactiplantibacillus plantarum, Bifidobacterium pseudocatenulatum and other combinations of probiotics capsules or tablets can useful as anti-inflammatory, anti-oxidant, or anti-pro inflammatory cytokines release and reduce the chances of occurring ND in patients [90]. Numerous neurological and psychiatric illnesses are linked to altered amounts of 5-HT (5-hydroxytryptamine-Serotonin) and DA (dopamine) to regulate this signaling molecule VSL#3 such as complex probiotic (consists of eight bacterial strains) VSL#3 interacts with mesenchymal stromal cells (hMSCs) to reduce neurodegeneration and inhibit NOD-like receptor protein-3, which mediated inflammation without altering the effects of hMSCs [91]. Cognitive processes, learning, memory, and emotional changes can all be modulated by NA. The two major inhibitory and excitatory chemicals are GABA and ACh. With respect to a single strain Bifidobacterium longum improve the cognitive function in healthy Balb/c mice, another context of using a multistrain probiotic, including different species of Lactobacilli and Bifidobacterium in the adult population, demonstrated improved cognition [92]. In accordance with the recent study to analyze the effect of a diet containing appropriate bacteria, participants were asked to consume two capsules after the meal in the morning and evening, which made a total of four capsules (a total of 1 × 109 colony-forming a unit of Bifidobacterium bifidum BGN4 and Bifidobacterium longum BORI in soybean oil) to be taken per day for 12 days and then the relative abundance at genus level of Clostridiales and Prevotellaceae has been observed [93].

4.5.1. Invertebrate System Studies

The beneficial effect of probiotics for neurodegenerative disease can be studied through an invertebrate model system, which is quite cheaper, and translational. Because of the complexity of the human nervous system and microbiota, identifying the primary microbial proteins or metabolites that have a direct influence on host neurons during neurodegeneration is usually difficult [94]. Simpler organisms like C. elegans were utilized in order to better understand the microbe-host interaction in the context of NDs. A recent study of the effect of probiotics on invertebrate models shows that, protein aggregation of α-synuclein, movement analysis, or locomotor analysis were restored with the help of a single bacterial strain (Lactobacillus plantarum) or multiple bacterial strains (E. coli OP50 and B. subtilis NCIB3610) [95].
In the PD model (C. elegans), expression of α-synuclein was expressed in disease the condition [96]. The Caldwell lab used a genome-wide RNAi screen to identify the role of the endocytic pathway in reducing α-synuclein toxicity in order to identify genetic variables that influence α-synuclein-mediated proteotoxicity and treated C. elegans with B. subtilis PB6 and Bifidobacterium dentium respectively that regulate the endocytic pathway through degradation of α-synuclein aggregates [97]. The PXN21 protein from B. subtilis prevents and prevents α-synuclein from aggregating [98]. The adult-onset loss of the dopaminergic neurons and locomotor dysfunction were induced by Ala-53-Thr, Ala-30-Pro, or Gln-46-Tyr mutations in the α-synuclein gene in the D. melanogaster variant of the UAS-Synuclein experiment, this locomotor dysfunction was restored by B. subtilis PXN21 and Lactobacillus plantarum within 4–12 days [99].
In the D. melanogaster strains of UAS-BACE/UAS-APP, L. brevis and Bifidobacterium dentium are less prevalent in the gut, and GABA levels are decreased in the CNS [100]. Similarly for AD model, uncoordinated locomotion, the buildup of insoluble tau, and age-related neuronal loss and deterioration are all present in C. elegans (A53T) mutant [101]. Although, Tan et al. found that Lactobacillus, particularly the L. plantarum DR7 strain, restores the rough eye phenotype in D. melanogaster GMR-A42 AD flies. The L. plantarum DR7 can restore the gut microbiota diversity in flies by increasing the abundance of Stenotrophomonas and Acetobacter, with reducing Wolbachia [102]. Although in other ND, like ALS; the generation of SOD1 (G85R) mutations in C. elegans led to severe locomotor defects and the formation of insoluble SOD1 aggregates in the perinuclear region of motor neurons [103]. In a demethylase-dependent manner, the KDM5 protein controls the immune deficiency (IMD) signaling pathway and maintains bacterial balance in D. melanogaster [104].

4.5.2. Vertebrate System Studies

The prevalence of ND is on the increase worldwide as the population ages, posing a serious danger to human health. Probiotics, live microorganisms that help the host’s health, may hold promise in the treatment and prevention of these crippling diseases, according to a recent study. Vertebrate models have become an important resource in this context for understanding the fundamental processes of neurodegeneration and evaluating the effectiveness of probiotics in reversing it. With respect to AD, according to Webberley et al.’s 2022 research in 3xTg mice, the Lab4b probiotic acts as a neuroprotective agent through an anti-inflammatory cytokine, and it has also been demonstrated that IL-10 absence lessens disease pathology in AD animals [105]. Similarly, Yang et al. discuss the importance of Acidophillus-KAL4 in reducing gut barrier damage and inflammation in elderly SAMP8 mouse models, as well as lowering levels of LPS and γ-H2AX, 8-OHdG, TLR4, RIG-I, and NF-κβ nuclear translocation in the brain [106]. In respect to PD, Sun et al. created male C57BL/6 (MPTP initiated) mice, and they investigated whether reversing gut microbiome dysbiosis was possible and Clostridium butyricum therapy for four weeks resulted in reduced amounts of colonic GLP-1, colonic GPR41/43, and cerebral GLP-1 receptor in MPTP-induced rodents [107]. By shedding light on the underlying mechanisms of these conditions and testing the efficacy of probiotics in vertebrate models, we might develop treatments that fight against these terrible illnesses Table 3.

4.6. Mechanism of Action and Therapeutic Effect of Probiotics in Combatting Neurological Disorders

Probiotics, which have been shown to have health benefits when consumed, have drawn a lot of interest in recent years because of their ability to treat and avoid a variety of diseases. While the exact mechanism of action of probiotics is not yet fully understood, growing evidence suggests that they act through various pathways to regulate immune function, improve gut barrier function, modulate the gut-brain axis and neurological complications [134].
Based on the results of both the animal and human studies, the consumption of probiotics has a significant beneficial effect on AD Table 3. We can conclude that most of the study is based on Bifidobacterium and Lactobacillus and as an outcome this study reveals that probiotics can improve memory dysfunction and cognitive dysfunction in similar to neurodegenerative diseases [87,98]. With respect to AD, the CNS’s inflammatory reaction to damage or infection is called neuroinflammation, which is accompanied by an accumulation of glial cells. Activated microglia and astrocytes generate pro-inflammatory cytokines like IL6, IL8, and IL10, and these cytokines directly cause neuronal injury [135]. According to studies, probiotics can restore chronic inflammation, the function of clearing abnormal proteins, and synaptic dysfunction. Neurodegeneration and brain loss are caused by all of these occurrences. To understand and solve the puzzle of how probiotics were beneficial in AD, there have primarily been four clinical trials, which are mentioned in Table 4, below. Regarding PD, a new clinical trial of probiotic capsules (containing Lactobacillus acidophilus, Bifidobacterium bifidum, Lactobacillus reuteri, and Lactobacillus fermentum) demonstrates the same effects as the MDS-UPDRS [136]. Another 2019 research demonstrates that the major pro- and anti-inflammatory cytokines, as well as ROS, are produced by Lactobacillus and Bifidobacterium genus when peripheral blood mononuclear cells (PBMCs) isolated from people with Parkinson’s disease (PD) are compared to healthy participants [137]. In relation to other ND Lacticaseibacillus rhamnosus HA-114 can improve the energy balance and cholesterol homeostasis in ND animals [131]. In relation to neurodegenerative disease, only 10% of the research concentrates on Streptococcus and Clostridium species. So on, we give a summary of the literature in this systematic review by using data that was extracted from different sources.

4.7. Future Aims and Conclusions

The use of probiotics in neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and other neurodegenerative disorders is an area of ongoing research, and there is not yet a definitive conclusion on its effectiveness. Some studies suggest that probiotics may have the potential in reducing inflammation and oxidative stress, improving gut microbiota composition, and enhancing cognitive function in neurodegenerative diseases. However, these findings are often based on invertebrate, animal models, or small-scale human studies, and more research is needed to confirm their efficacy and safety. It is important to note that probiotics should not be considered a cure or a standalone treatment for neurodegenerative diseases. They should be used as part of a comprehensive treatment plan, including medication, lifestyle changes, and other interventions recommended by healthcare professionals. Overall, while the use of Bifidobacterium and Lactobacillus in neurodegenerative diseases is a promising area of research, more large-scale, randomized controlled trials are needed to better understand their potential benefits and limitations.

Author Contributions

Conceptualization, M.J.; methodology, N.P.; software, N.P.; validation, M.J., C.K. and P.K.; formal analysis, M.J.; data curation, S.O.; writing—original draft preparation, N.P. and S.O.; writing—review and editing M.J. and P.K.; supervision, C.K. and M.J.; project administration, P.K. and C.K.; funding acquisition, P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study is funded by Young Scientist grant, Human Resource Development Scheme of Department of Health and Research, Ministry of health and family welfare, Government of India and ICMR (YSS/2020/000113/PRCYSS) and Intramural Research grant, Parul University (CR4D/IMSL/048).

Data Availability Statement

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jellinger, K.A. Basic mechanisms of neurodegeneration: A critical update. J. Cell. Mol. Med. 2010, 14, 457–487. [Google Scholar] [CrossRef]
  2. Brown, R.C.; Lockwood, A.H.; Sonawane, B.R. Neurodegenerative Diseases: An Overview of Environmental Risk Factors. Environ. Health Perspect. 2005, 113, 1250–1256. [Google Scholar] [CrossRef]
  3. Kechagia, M.; Basoulis, D.; Konstantopoulou, S.; Dimitriadi, D.; Gyftopoulou, K.; Skarmoutsou, N.; Fakiri, E.M. Health Benefits of Probiotics: A Review. ISRN Nutr. 2013, 2013, 481651. [Google Scholar] [CrossRef] [PubMed]
  4. Valdes, A.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef]
  5. Silva, D.R.; Sardi, J.D.; de Souza Pitangui, N.; Roque, S.M.; da Silva, A.C.; Rosalen, P.L. Probiotics as an alternative antimicrobial therapy: Current reality and future directions. J. Funct. Foods 2020, 73, 104080. [Google Scholar] [CrossRef]
  6. Sun, P.; Su, L.; Zhu, H.; Li, X.; Guo, Y.; Du, X.; Zhang, L.; Qin, C. Gut Microbiota Regulation and Their Implication in the Development of Neurodegenerative Disease. Microorganisms 2021, 9, 2281. [Google Scholar] [CrossRef] [PubMed]
  7. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
  8. Drago, L.O.; De Vecchi, E. Should Lactobacillus sporogenes and Bacillus coagulans have a future? J. Chemother. 2009, 21, 371–377. [Google Scholar] [CrossRef] [PubMed]
  9. Cortese, Y.J.; Wagner, V.E.; Tierney, M.; Devine, D.; Fogarty, A. Review of Catheter-Associated Urinary Tract Infections and In Vitro Urinary Tract Models. J. Healthc. Eng. 2018, 2018, 2986742. [Google Scholar] [CrossRef]
  10. Doron, S.; Snydman, D.R. Risk and Safety of Probiotics. Clin. Infect. Dis. 2015, 60 (Suppl. S2), S129–S134. [Google Scholar] [CrossRef]
  11. Bathina, S.; Das, U.N. Brain-derived neurotrophic factor and its clinical implications. Arch. Med. Sci. 2015, 11, 1164–1178. [Google Scholar] [CrossRef]
  12. Ahn, E.; Kang, H. Introduction to systematic review and meta-analysis. Korean J. Anesthesiol. 2018, 71, 103–112. [Google Scholar] [CrossRef] [PubMed]
  13. Ruiz-Gonzalez, C.; Roman, P.; Rueda-Ruzafa, L.; Rodriguez-Arrastia, M.; Cardona, D. Effects of probiotics supplementation on dementia and cognitive impairment: A systematic review and meta-analysis of preclinical and clinical studies. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 108, 110189. [Google Scholar] [CrossRef]
  14. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Syst. Rev. 2021, 89, 105906. [Google Scholar]
  15. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Park. Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
  16. Higgins, J.P.T.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef]
  17. Saulnier, D.M.; Ringel, Y.; Heyman, M.B.; Foster, J.A.; Bercik, P.; Shulman, R.J.; Versalovic, J.; Verdu, E.F.; Dinan, T.G.; Hecht, G.; et al. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology. Gut Microbes 2013, 4, 17–27. [Google Scholar] [CrossRef]
  18. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-Gut-Microbe Communication in Health and Disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef] [PubMed]
  19. Alkasir, R.; Li, J.; Li, X.; Jin, M.; Zhu, B. Human gut microbiota: The links with dementia development. Protein Cell 2017, 8, 90–102. [Google Scholar] [CrossRef]
  20. Hawkes, L.A.; Broderick, A.C.; Godfrey, M.H.; Godley, B.J. Investigating the potential impacts of climate change on a marine turtle population. Glob. Chang. Biol. 2007, 13, 923–932. [Google Scholar] [CrossRef]
  21. Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef]
  22. Dando, S.J.; Mackay-Sim, A.; Norton, R.; Currie, B.J.; John, J.A.S.; Ekberg, J.A.K.; Batzloff, M.; Ulett, G.C.; Beacham, I.R. Pathogens Penetrating the Central Nervous System: Infection Pathways and the Cellular and Molecular Mechanisms of Invasion. Clin. Microbiol. Rev. 2014, 27, 691–726. [Google Scholar] [CrossRef]
  23. Zhang, H.; Chen, Y.; Wang, Z.; Xie, G.; Liu, M.; Yuan, B.; Chai, H.; Wang, W.; Cheng, P. Implications of gut microbiota in neurodegenerative diseases. Front. Immunol. 2022, 13, 325. [Google Scholar] [CrossRef] [PubMed]
  24. Fedeli, C.; Filadi, R.; Rossi, A.; Mammucari, C.; Pizzo, P. PSEN2 (presenilin 2) mutants linked to familial Alzheimer disease impair autophagy by altering Ca2+ homeostasis. Autophagy 2019, 15, 2044–2062. [Google Scholar] [CrossRef] [PubMed]
  25. Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef] [PubMed]
  26. Rieder, R.; Wisniewski, P.J.; Alderman, B.L.; Campbell, S.C. Microbes and mental health: A review. Brain Behav. Immun. 2017, 66, 9–17. [Google Scholar] [CrossRef]
  27. Nagahara, A.H.; Tuszynski, M.H. Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat. Rev. Drug Discov. 2011, 10, 209–219. [Google Scholar] [CrossRef]
  28. Bell, K.F.; Hardingham, G.E. The influence of synaptic activity on neuronal health. Curr. Opin. Neurobiol. 2011, 21, 299–305. [Google Scholar] [CrossRef]
  29. Perez, S.E.; Lazarov, O.; Koprich, J.B.; Chen, E.Y.; Rodriguez-Menendez, V.; Lipton, J.W.; Sisodia, S.S.; Mufson, E.J. Nigrostriatal dysfunction in familial Alzheimer’s disease-linked APPswe/PS1ΔE9 transgenic mice. J. Neurosci. 2005, 25, 10220–10229. [Google Scholar] [CrossRef]
  30. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef]
  31. Wu, C.; Sun, D. GABA receptors in brain development, function, and injury. Metab. Brain Dis. 2015, 30, 367–379. [Google Scholar] [CrossRef]
  32. Kesika, P.; Suganthy, N.; Sivamaruthi, B.S.; Chaiyasut, C. Role of gut-brain axis, gut microbial composition, and probiotic intervention in Alzheimer’s disease. Life Sci. 2021, 264, 118627. [Google Scholar] [CrossRef]
  33. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [PubMed]
  34. Roubaud-Baudron, C.; Krolak-Salmon, P.; Quadrio, I.; Mégraud, F.; Salles, N. Impact of chronic Helicobacter pylori infection on Alzheimer’s disease: Preliminary results. Neurobiol. Aging 2012, 33, 1009.e11–1009.e19. [Google Scholar] [CrossRef] [PubMed]
  35. Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef]
  36. Fasano, A.; Bove, F.; Gabrielli, M.; Petracca, M.; Zocco, M.A.; Ragazzoni, E.; Barbaro, F.; Piano, C.; Fortuna, S.; Tortora, A.; et al. The role of small intestinal bacterial overgrowth in Parkinson’s disease. Mov. Disord. 2013, 28, 1241–1249. [Google Scholar] [CrossRef]
  37. Tan, A.H.; Mahadeva, S.; Thalha, A.M.; Gibson, P.R.; Kiew, C.K.; Yeat, C.M.; Ng, S.W.; Ang, S.P.; Chow, S.K.; Tan, C.T.; et al. Small intestinal bacterial overgrowth in Parkinson’s disease. Park. Relat. Dis. 2014, 20, 535–540. [Google Scholar] [CrossRef]
  38. Derkinderen, P.; Shannon, K.M.; Brundin, P. Gut feelings about smoking and coffee in Parkinson’s disease. Mov. Disord. 2014, 29, 976–979. [Google Scholar] [CrossRef]
  39. Schaeffer, E.; Kluge, A.; Böttner, M.; Zunke, F.; Cossais, F.; Berg, D.; Arnold, P. Alpha synuclein connects the gut-brain axis in Parkinson’s disease patients–a view on clinical aspects, cellular pathology and analytical methodology. Front. Cell Dev. Biol. 2020, 8, 573696. [Google Scholar] [CrossRef]
  40. Du, G.; Dong, W.; Yang, Q.; Yu, X.; Ma, J.; Gu, W.; Huang, Y. Altered Gut Microbiota Related to Inflammatory Responses in Patients with Huntington’s Disease. Front. Immunol. 2021, 11, 603594. [Google Scholar] [CrossRef] [PubMed]
  41. Kong, G.; Ellul, S.; Narayana, V.K.; Kanojia, K.; Ha, H.T.; Li, S.; Renoir, T.; Le Cao, K.A.; Hannan, A.J. An integrated metagenomics and metabolomics approach implicates the microbiota-gut-brain axis in the pathogenesis of Huntington’s disease. Neurobiol. Dis. 2021, 148, 105199. [Google Scholar] [CrossRef] [PubMed]
  42. Ferrante, R.J.; Andreassen, O.A.; Jenkins, B.G.; Dedeoglu, A.; Kuemmerle, S.; Kubilus, J.K.; Kaddurah-Daouk, R.; Hersch, S.M.; Beal, M.F. Neuroprotective effects of creatine in a transgenic mouse model of Huntington’s disease. J. Neurosci. 2000, 20, 4389–4397. [Google Scholar] [CrossRef]
  43. van der Burg, J.M.; Winqvist, A.; Aziz, N.A.; Maat-Schieman, M.L.; Roos, R.A.; Bates, G.P.; Brundin, P.; Björkqvist, M.; Wierup, N. Gastrointestinal dysfunction contributes to weight loss in Huntington’s disease mice. Neurobiol. Dis. 2011, 44, 1–8. [Google Scholar] [CrossRef] [PubMed]
  44. Marć, M.A.; Jastrząb, R.; Mytych, J. Does the Gut Microbial Metabolome Really Matter? The Connection between GUT Metabolome and Neurological Disorders. Nutrients 2022, 14, 3967. [Google Scholar] [CrossRef]
  45. Gubert, C.; Choo, J.M.; Love, C.J.; Kodikara, S.; Masson, B.A.; Liew, J.J.M.; Wang, Y.; Kong, G.; Narayana, V.K.; Renoir, T. Faecal microbiota transplant ameliorates gut dysbiosis and cognitive deficits in Huntington’s disease mice. Brain Commun. 2022, 4, fcac205. [Google Scholar] [CrossRef]
  46. Cai, Z.; Hussain, M.D.; Yan, L.J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 2014, 124, 307–321. [Google Scholar] [CrossRef]
  47. Round, J.L.; Mazmanian, S.K. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 2009, 9, 313–323. [Google Scholar] [CrossRef]
  48. Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 359, 1151–1156. [Google Scholar] [CrossRef] [PubMed]
  49. Chio, A.; Logroscino, G.; Hardiman, O.; Swingler, R.; Mitchell, D.; Beghi, E.; Traynor, B.; On Behalf of the Eurals Consortium. Prognostic factors in ALS: A critical review. ALS 2009, 10, 310–323. [Google Scholar] [CrossRef]
  50. Rowin, J.; Xia, Y.; Jung, B.; Sun, J. Gut inflammation and dysbiosis in human motor neuron disease. Physiol. Rep. 2017, 5, e13443. [Google Scholar] [CrossRef]
  51. Brenner, D.; Hiergeist, A.; Adis, C.; Mayer, B.; Gessner, A.; Ludolph, A.C.; Weishaupt, J.H. The fecal microbiome of ALS patients. Neurobiol. Aging 2018, 61, 132–137. [Google Scholar] [CrossRef] [PubMed]
  52. Houser, M.C.; Tansey, M.G. The gut-brain axis: Is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? npj Park. Dis. 2017, 3, 3. [Google Scholar] [CrossRef]
  53. Wolf Susanne, A.; Boddeke, H.W.; Kettenmann, H. Microglia in physiology and disease. Annu. Rev. Physiol. 2017, 79, 619–643. [Google Scholar] [CrossRef] [PubMed]
  54. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed]
  55. Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar]
  56. Kaur, H.; Bose, C.; Mande, S.S. Tryptophan Metabolism by Gut Microbiome and Gut-Brain-Axis: An in silico Analysis. Front. Neurosci. 2019, 13, 1365. [Google Scholar] [CrossRef]
  57. Bonaz, B.; Bazin, T.; Pellissier, S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front. Neurosci. 2018, 12, 49. [Google Scholar] [CrossRef]
  58. Del Tredici, K.; Braak, H. Sporadic Parkinson’s disease: Development and distribution of α-synuclein pathology. Neuropathol. Appl. Neurobiol. 2016, 42, 33–50. [Google Scholar] [CrossRef]
  59. Versalovic, J. The Human Microbiome and Probiotics: Implications for Pediatrics. Ann. Nutr. Metab. 2013, 63 (Suppl. S2), 42–52. [Google Scholar] [CrossRef]
  60. Kamada, N.; Chen, G.Y.; Inohara, N.; Núñez, G. Control of pathogens and pathobionts by the gut microbiota. Nat. Immunol. 2013, 14, 685–690. [Google Scholar] [CrossRef]
  61. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [PubMed]
  62. Effendi, R.M.R.A.; Anshory, M.; Kalim, H.; Dwiyana, R.F.; Suwarsa, O.; Pardo, L.M.; Nijsten, T.E.C.; Thio, H.B. Akkermansia muciniphila and Faecalibacterium prausnitzii in Immune-Related Diseases. Microorganisms 2022, 10, 2382. [Google Scholar] [CrossRef] [PubMed]
  63. Bo, T.-B.; Wen, J.; Zhao, Y.-C.; Tian, S.-J.; Zhang, X.-Y.; Wang, D.-H. Bifidobacterium pseudolongum reduces triglycerides by modulating gut microbiota in mice fed high-fat food. J. Steroid Biochem. Mol. Biol. 2020, 198, 105602. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, X.; Zhang, P.; Zhang, X. Probiotics Regulate Gut Microbiota: An Effective Method to Improve Immunity. Molecules 2021, 26, 6076. [Google Scholar] [CrossRef]
  65. Johansson, M.E.V.; Hansson, G.C. Immunological aspects of intestinal mucus and mucins. Nat. Rev. Immunol. 2016, 16, 639–649. [Google Scholar] [CrossRef]
  66. Trop, T.K. Intestinal microbiota, probiotics and prebiotics in inflammatory bowel disease. World J. Gastroenterol. WJG 2014, 20, 11505. [Google Scholar]
  67. Satoh, T.; Izumi, H.; Iwabuchi, N.; Odamaki, T.; Namba, K.; Abe, F.; Xiao, J. Bifidobacterium breve prevents necrotising enterocolitis by suppressing inflammatory responses in a preterm rat model. Benef. Microbes 2016, 7, 75–82. [Google Scholar] [CrossRef]
  68. Aloisio, I.; Prodam, F.; Giglione, E.; Bozzi Cionci, N.; Solito, A.; Bellone, S.; Baffoni, L.; Mogna, L.; Pane, M.; Bona, G.; et al. Three-Month Feeding Integration With Bifidobacterium Strains Prevents Gastrointestinal Symptoms in Healthy Newborns. Front. Nutr. 2018, 5, 39. [Google Scholar] [CrossRef]
  69. Leech, J.; Cabrera-Rubio, R.; Walsh, A.M.; Macori, G.; Walsh, C.J.; Barton, W.; Finnegan, L.; Crispie, F.; O’sullivan, O.; Claesson, M.J.; et al. Fermented-Food Metagenomics Reveals Substrate-Associated Differences in Taxonomy and Health-Associated and Antibiotic Resistance Determinants. Msystems 2020, 5, e00522-20. [Google Scholar] [CrossRef]
  70. Yao, S.; Zhao, Z.; Wang, W.; Liu, X. Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J. Immunol. Res. 2021, 2021, 8030297. [Google Scholar] [CrossRef]
  71. Moloney, G.M.; Long-Smith, C.M.; Murphy, A.; Dorland, D.; Hojabri, S.F.; Ramirez, L.O.; Marin, D.C.; Bastiaanssen, T.F.; Cusack, A.-M.; Berding, K.; et al. Improvements in sleep indices during exam stress due to consumption of a Bifidobacterium longum. Brain Behav. Immun.-Health 2021, 10, 100174. [Google Scholar] [CrossRef] [PubMed]
  72. Schell, M.A.; Karmirantzou, M.; Snel, B.; Vilanova, D.; Berger, B.; Pessi, G.; Zwahlen, M.-C.; Desiere, F.; Bork, P.; Delley, M.; et al. The genome sequence of Bifidobacterium longum reflects its adaptation to the human gastrointestinal tract. Proc. Natl. Acad. Sci. USA 2002, 99, 14422–14427. [Google Scholar] [CrossRef] [PubMed]
  73. Aoki, R.; Tsuchida, S.; Arai, Y.; Ohno, K.; Nishijima, T.; Mawatari, T.; Mikami, Y.; Ushida, K. Effect of Bifidobacterium animalis subsp. lactis GCL2505 on the physiological function of intestine in a rat model. Food Sci. Nutr. 2016, 4, 782–790. [Google Scholar] [CrossRef] [PubMed]
  74. Daelemans, S.; Peeters, L.; Hauser, B.; Vandenplas, Y. Recent advances in understanding and managing infantile colic. F1000Research 2018, 7, 1426. [Google Scholar] [CrossRef] [PubMed]
  75. Jungersen, M.; Wind, A.; Johansen, E.; Christensen, J.E.; Stuer-Lauridsen, B.; Eskesen, D. The Science behind the Probiotic Strain Bifidobacterium animalis subsp. lactis BB-12®. Microorganisms 2014, 2, 92–110. [Google Scholar] [CrossRef]
  76. Rossi, M.; Amaretti, A.; Raimondi, S. Folate Production by Probiotic Bacteria. Nutrients 2011, 3, 118–134. [Google Scholar] [CrossRef]
  77. Kaźmierczak-Siedlecka, K.; Roviello, G.; Catalano, M.; Polom, K. Gut microbiota modulation in the context of immune-related aspects of Lactobacillus spp. and Bifidobacterium spp. in gastrointestinal cancers. Nutrients 2021, 13, 2674. [Google Scholar] [CrossRef]
  78. Chen, Y.; Yang, B.; Stanton, C.; Ross, R.P.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium pseudocatenulatum ameliorates DSS-induced colitis by maintaining intestinal mechanical barrier, blocking proinflammatory cytokines, inhibiting TLR4/NF-κB signaling, and altering gut microbiota. J. Agric. Food Chem. 2021, 69, 1496–1512. [Google Scholar] [CrossRef]
  79. Yang, X.; Lu, D.; Zhuo, J.; Lin, Z.; Yang, M.; Xu, X. The Gut-liver Axis in Immune Remodeling: New insight into Liver Diseases. Int. J. Biol. Sci. 2020, 16, 2357–2366. [Google Scholar] [CrossRef]
  80. Geerlings, S.Y.; Kostopoulos, I.; De Vos, W.M.; Belzer, C. Akkermansia muciniphila in the Human Gastrointestinal Tract: When, Where, and How? Microorganisms 2018, 6, 75. [Google Scholar] [CrossRef]
  81. Rodrigues, V.F.; Elias-Oliveira, J.; Pereira, S.; Pereira, J.A.; Barbosa, S.C.; Machado, M.S.G.; Carlos, D. Akkermansia muciniphila and Gut Immune System: A Good Friendship That Attenuates Inflammatory Bowel Disease, Obesity, and Diabetes. Front. Immunol. 2022, 13, 934695. [Google Scholar] [CrossRef] [PubMed]
  82. Kalia, V.C.; Gong, C.; Shanmugam, R.; Lin, H.; Zhang, L.; Lee, J.-K. The Emerging Biotherapeutic Agent: Akkermansia. Indian J. Microbiol. 2022, 62, 1–10. [Google Scholar] [CrossRef]
  83. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef]
  84. Śliżewska, K.; Markowiak-Kopeć, P.; Śliżewska, W. The role of probiotics in cancer prevention. Cancers 2020, 13, 20. [Google Scholar] [CrossRef] [PubMed]
  85. Bertuccini, L.; Russo, R.; Iosi, F.; Superti, F. Effects of Lactobacillus rhamnosus and Lactobacillus acidophilus on bacterial vaginal pathogens. Int. J. Immunopathol. Pharmacol. 2017, 30, 163–167. [Google Scholar] [CrossRef] [PubMed]
  86. Bermúdez-Humarán, L.G.; Innocentin, S.; Lefèvre, F.; Chatel, J.-M.; Langella, P. Development of Mucosal Vaccines Based on Lactic Acid Bacteria. Prebiotics Probiotics Sci. Technol. 2009. [Google Scholar] [CrossRef]
  87. Jaradat, N.; Khasati, A.; Hawi, M.; Hawash, M.; Shekfeh, S.; Qneibi, M.; Eid, A.M.; Arar, M.; Qaoud, M.T. Antidiabetic, antioxidant, and anti-obesity effects of phenylthio-ethyl benzoate derivatives, and molecular docking study regarding α-amylase enzyme. Sci. Rep. 2022, 12, 3108. [Google Scholar] [CrossRef]
  88. Zhang, X.; Hu, C.; Zhong, Y.; Qiao, D.; Chi, W.; Shen, H.; Chong, W. Multifunctional Interleukin-24 Resolves Neuroretina Auto-immunity via Diverse Mechanisms. Int. J. Mol. Sci. 2022, 23, 11988. [Google Scholar] [CrossRef]
  89. Fu, J.; Wang, T.; Xiao, X.; Cheng, Y.; Wang, F.; Jin, M.; Wang, Y.; Zong, X. Clostridium Butyricum ZJU-F1 Benefits the Intestinal Barrier Function and Immune Response Associated with Its Modulation of Gut Microbiota in Weaned Piglets. Cells 2021, 10, 527. [Google Scholar] [CrossRef]
  90. Eifler, N.; Vetsch, M.; Gregorini, M.; Ringler, P.; Chami, M.; Philippsen, A.; Fritz, A.; Müller, S.A.; Glockshuber, R.; Engel, A.; et al. Cytotoxin ClyA from Escherichia coli assembles to a 13-meric pore independent of its redox-state. EMBO J. 2006, 25, 2652–2661. [Google Scholar] [CrossRef]
  91. Pajares, M.; IRojo, A.; Manda, G.; Boscá, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells 2020, 9, 1687. [Google Scholar] [CrossRef]
  92. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  93. Kim, C.-S.; Cha, L.; Sim, M.; Jung, S.; Chun, W.Y.; Baik, H.W.; Shin, D.-M. Probiotic Supplementation Improves Cognitive Function and Mood with Changes in Gut Microbiota in Community-Dwelling Older Adults: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. J. Gerontol. Ser. A 2021, 76, 32–40. [Google Scholar] [CrossRef] [PubMed]
  94. Zhou, L.; Han, D.; Wang, X.; Chen, Z. Probiotic Formulation VSL# 3 Interacts with Mesenchymal Stromal Cells to Protect Dopaminergic Neurons via Centrally and Peripherally Suppressing NOD-Like Receptor Protein 3 Inflammasome-Mediated Inflammation in Parkinson’s Disease Mice. Microbiol. Spectr. 2023, 2, e03208-22. [Google Scholar]
  95. Savignac, H.M.; Kiely, B.; Dinan, T.G.; Cryan, J.F. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol. Motil. 2014, 26, 1615–1627. [Google Scholar] [CrossRef]
  96. Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef]
  97. Nussbaum-Krammer, C.I.; Morimoto, R.I. Caenorhabditis elegans as a model system for studying non-cell-autonomous mechanisms in protein-misfolding diseases. Dis. Model. Mech. 2014, 7, 31–39. [Google Scholar] [CrossRef]
  98. Gaeta, A.L.; Caldwell, K.A.; Caldwell, G.A. Found in translation: The utility of C. elegans alpha-synuclein models of Parkinson’s disease. Brain Sci. 2019, 9, 73. [Google Scholar] [CrossRef] [PubMed]
  99. Gitler, A.D.; Chesi, A.; Geddie, M.L.; Strathearn, K.E.; Hamamichi, S.; Hill, K.J.; Caldwell, K.A.; Caldwell, G.A.; Cooper, A.A.; Rochet, J.-C.; et al. α-Synuclein is part of a diverse and highly conserved interaction network that includes PARK9 and manganese toxicity. Nat. Genet. 2009, 41, 308–315. [Google Scholar] [CrossRef]
  100. Goya, M.E.; Xue, F.; Sampedro-Torres-Quevedo, C.; Arnaouteli, S.; Riquelme-Dominguez, L.; Romanowski, A.; Brydon, J.; Ball, K.L.; Stanley-Wall, N.R.; Doitsidou, M. Probiotic Bacillus subtilis protects against α-synuclein aggregation in C. elegans. Cell Rep. 2020, 30, 367–380. [Google Scholar] [CrossRef]
  101. Reiszadeh Jahromi, S.; Ramesh, S.R.; Finkelstein, D.I.; Haddadi, M. α-synuclein E46K mutation and involvement of oxidative stress in a drosophila model of Parkinson’s disease. Park. Dis. 2021, 2021, 6621507. [Google Scholar] [CrossRef] [PubMed]
  102. Pokusaeva, K.; Johnson, C.; Luk, B.; Uribe, G.; Fu, Y.; Oezguen, N.; Matsunami, R.K.; Lugo, M.; Major, A.; Mori-Akiyama, Y.; et al. GABA-producing Bifidobacterium dentium modulates visceral sensitivity in the intestine. Neurogastroenterol. Motil. 2017, 29, e12904. [Google Scholar] [CrossRef] [PubMed]
  103. Van Pelt, K.M.; Truttmann, M.C. Caenorhabditis elegans as a model system for studying aging-associated neurodegenerative diseases. Transl. Med. Aging 2020, 4, 60–72. [Google Scholar] [CrossRef]
  104. Tan, F.H.; Liu, G.; Lau, S.Y.; Jaafar, M.H.; Park, Y.H.; Azzam, G.; Li, Y.; Liong, M.T. Lactobacillus probiotics improved the gut microbiota profile of a Drosophila melanogaster Alzheimer’s disease model and alleviated neurodegeneration in the eye. Benef. Microbes 2020, 11, 79–89. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, J.; Farr, G.W.; Hall, D.H.; Li, F.; Furtak, K.; Dreier, L.; Horwich, A.L. An ALS-Linked Mutant SOD1 Produces a Locomotor Defect Associated with Aggregation and Synaptic Dysfunction When Expressed in Neurons of Caenorhabditis elegans. PLoS Genet. 2009, 5, e1000350. [Google Scholar] [CrossRef]
  106. Chen, K.; Luan, X.; Liu, Q.; Wang, J.; Chang, X.; Snijders, A.M.; Mao, J.-H.; Secombe, J.; Dan, Z.; Chen, J.-H.; et al. Drosophila Histone Demethylase KDM5 Regulates Social Behavior through Immune Control and Gut Microbiota Maintenance. Cell Host Microbe 2019, 25, 537–552.e8. [Google Scholar] [CrossRef]
  107. Webberley, T.S.; Bevan, R.J.; Kerry-Smith, J.; Dally, J.; Michael, D.R.; Thomas, S.; Rees, M.; Morgan, J.E.; Marchesi, J.R.; Good, M.A.; et al. Assessment of Lab4P Probiotic Effects on Cognition in 3xTg-AD Alzheimer’s Disease Model Mice and the SH-SY5Y Neuronal Cell Line. Int. J. Mol. Sci. 2023, 24, 4683. [Google Scholar] [CrossRef]
  108. Yang, Y.J.; Chuang, C.C.; Yang, H.B.; Lu, C.C.; Sheu, B.S. Lactobacillus acidophilus ameliorates H. pylori-induced gastric inflammation by inactivating the Smad7 and NFκB pathways. BMC Microbiol. 2012, 12, 38. [Google Scholar] [CrossRef]
  109. Sun, J.; Wang, F.; Hu, X.; Yang, C.; Xu, H.; Yao, Y.; Liu, J. Clostridium butyricum Attenuates Chronic Unpredictable Mild Stress-Induced Depressive-Like Behavior in Mice via the Gut-Brain Axis. J. Agric. Food Chem. 2018, 66, 8415–8421. [Google Scholar] [CrossRef]
  110. Varesi, A.; Campagnoli, L.I.M.; Fahmideh, F.; Pierella, E.; Romeo, M.; Ricevuti, G.; Nicoletta, M.; Chirumbolo, S.; Pascale, A. The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment. Int. J. Mol. Sci. 2022, 23, 12289. [Google Scholar] [CrossRef]
  111. Cogliati, S.; Clementi, V.; Francisco, M.; Crespo, C.; Arganaraz, F.; Grau, R. Bacillus subtilis delays neurodegeneration and behavioral impairment in the Alzheimer’s disease model Caenorhabditis elegans. J. Alzheimer’s Dis. 2020, 73, 1035–1052. [Google Scholar] [CrossRef] [PubMed]
  112. Labarre, A.; Guitard, E.; Tossing, G.; Bareke, E.; Labrecque, M.; Tetreault, M.; Parker, A. Probiotic Lacticaseibacillus rhamnosus HA-114 Suppresses Age-Dependent Neurodegeneration via Mitochondrial Beta-Oxidation. Res. Sq. 2020. [Google Scholar] [CrossRef]
  113. Donato, V.; Ayala, F.R.; Cogliati, S.; Bauman, C.; Costa, J.G.; Leñini, C.; Grau, R. Bacillus subtilis biofilm extends Caenorhabditis elegans longevity through downregulation of the insulin-like signalling pathway. Nat. Commun. 2017, 8, 14332. [Google Scholar] [CrossRef]
  114. Zaydi, A.I.; Lew, L.C.; Hor, Y.Y.; Jaafar, M.H.; Chuah, L.O.; Yap, K.P.; Azlan, A.; Azzam, G.; Liong, M.T. Lactobacillus plantarum DR7 improved brain health in aging rats via the serotonin, inflammatory and apoptosis pathways. Benef. Microbes 2020, 11, 753–766. [Google Scholar] [CrossRef] [PubMed]
  115. Ruiz, L.; Flórez, A.B.; Sánchez, B.; Moreno-Muñoz, J.A.; Rodriguez-Palmero, M.; Jiménez, J.; Gavilán, C.G.d.l.R.; Gueimonde, M.; Ruas-Madiedo, P.; Margolles, A. Bifidobacterium longum subsp. infantis CECT7210 (B. infantis IM-1®) Displays In Vitro Activity against Some Intestinal Pathogens. Nutrients 2020, 12, 3259. [Google Scholar] [CrossRef] [PubMed]
  116. Salles, B.I.M.; Cioffi, D.; Ferreira, S.R.G. Probiotics supplementation and insulin resistance: A systematic review. Diabetol. Metab. Syndr. 2020, 12, 98. [Google Scholar] [CrossRef] [PubMed]
  117. Saini, N.; Georgiev, O.; Schaffner, W. The parkin Mutant Phenotype in the Fly Is Largely Rescued by Metal-Responsive Transcription Factor (MTF-1). Mol. Cell. Biol. 2011, 31, 2151–2161. [Google Scholar] [CrossRef]
  118. Giles, H.H.; Petersen, S.C.; Call, G.B.; Chaston, J.M. Genome Sequence of Lactiplantibacillus plantarum DmPark25_157, a Bacterial Strain Isolated from Drosophila melanogaster. Microbiol. Resour. Announc. 2021, 10, e01372-20. [Google Scholar] [CrossRef]
  119. Chongtham, A.; Yoo, J.H.; Chin, T.M.; Akingbesote, N.D.; Huda, A.; Marsh, J.L.; Khoshnan, A. Gut Bacteria Regulate the Pathogenesis of Huntington’s Disease in Drosophila Model. Front. Neurosci. 2022, 16, 902205. [Google Scholar] [CrossRef]
  120. Huang, H.J.; Chen, J.L.; Liao, J.F.; Chen, Y.H.; Chieu, M.W.; Ke, Y.Y.; Hsu, C.C.; Tsai, Y.C.; Hsieh-Li, H.M. Lactobacillus plantarum PS128 prevents cognitive dysfunction in Alzheimer’s disease mice by modulating propionic acid levels, glycogen synthase kinase 3 beta activity, and gliosis. BMC Complement. Med. Ther. 2021, 21, 259. [Google Scholar] [CrossRef]
  121. Abdelhamid, M.; Zhou, C.; Ohno, K.; Kuhara, T.; Taslima, F.; Abdullah, M.; Jung, C.-G.; Michikawa, M. Probiotic Bifidobacterium breve Prevents Memory Impairment Through the Reduction of Both Amyloid-β Production and Microglia Activation in APP Knock-In Mouse. J. Alzheimer’s Dis. 2022, 85, 1555–1571. [Google Scholar] [CrossRef]
  122. Suire, C.N.; Abdul-Hay, S.O.; Sahara, T.; Kang, D.; Brizuela, M.K.; Saftig, P.; Dickson, D.W.; Rosenberry, T.L.; Leissring, M.A. Cathepsin D regulates cerebral Aβ42/40 ratios via differential degradation of Aβ42 and Aβ40. Alzheimer’s Res. Ther. 2020, 12, 80. [Google Scholar] [CrossRef] [PubMed]
  123. Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef]
  124. Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, 10428. [Google Scholar] [CrossRef] [PubMed]
  125. Wong, C.B.; Iwabuchi, N.; Xiao, J.-Z. Exploring the Science behind Bifidobacterium breve M-16V in Infant Health. Nutrients 2019, 11, 1724. [Google Scholar] [CrossRef]
  126. Enomoto, T.; Sowa, M.; Nishimori, K.; Shimazu, S.; Yoshida, A.; Yamada, K.; Furukawa, F.; Nakagawa, T.; Yanagisawa, N.; Iwabuchi, N.; et al. Effects of Bifidobacterial Supplementation to Pregnant Women and Infants in the Prevention of Allergy Development in Infants and on Fecal Microbiota. Allergol. Int. 2014, 63, 575–585. [Google Scholar] [CrossRef] [PubMed]
  127. Han, Y.; Kim, B.; Ban, J.; Lee, J.; Kim, B.J.; Choi, B.S.; Hwang, S.; Ahn, K.; Kim, J. A randomized trial of Lactobacillus plantarum CJLP 133 for the treatment of atopic dermatitis. Pediatr. Allergy Immunol. 2012, 23, 667–673. [Google Scholar] [CrossRef]
  128. Matsumoto, M.; Aranami, A.; Ishige, A.; Watanabe, K.; Benno, Y. LKM512 yogurt consumption improves the intestinal environment and induces the T-helper type 1 cytokine in adult patients with intractable atopic dermatitis. Clin. Exp. Allergy 2007, 37, 358–370. [Google Scholar] [CrossRef]
  129. Hsieh, T.-H.; Kuo, C.-W.; Hsieh, K.-H.; Shieh, M.-J.; Peng, C.-W.; Chen, Y.-C.; Chang, Y.-L.; Huang, Y.-Z.; Chen, C.-C.; Chang, P.-K.; et al. Probiotics Alleviate the Progressive Deterioration of Motor Functions in a Mouse Model of Parkinson’s Disease. Brain Sci. 2020, 10, 206. [Google Scholar] [CrossRef]
  130. Ishii, T.; Furuoka, H.; Kaya, M.; Kuhara, T. Oral Administration of Probiotic Bifidobacterium breve Improves Facilitation of Hippocampal Memory Extinction via Restoration of Aberrant Higher Induction of Neuropsin in an MPTP-Induced Mouse Model of Parkinson’s Disease. Biomedicines 2021, 9, 167. [Google Scholar] [CrossRef]
  131. Tytgat, H.L.; Douillard, F.P.; Reunanen, J.; Rasinkangas, P.; Hendrickx, A.P.; Laine, P.K.; Paulin, L.; Satokari, R.; de Vos, W.M. Lactobacillus rhamnosus GG Outcompetes Enterococcus faecium via Mu-cus-Binding Pili: Evidence for a Novel and Heterospecific Probiotic Mechanism. Appl. Environ. Microbiol. 2016, 82, 5756–5762. [Google Scholar] [CrossRef]
  132. Labarre, A.; Guitard, E.; Tossing, G.; Forest, A.; Bareke, E.; Labrecque, M.; Tétreault, M.; Ruiz, M.; Parker, J.A. Fatty acids derived from the probiotic Lacticaseibacillus rhamnosus HA-114 suppress age-dependent neurodegeneration. Commun. Biol. 2022, 5, 1340. [Google Scholar] [CrossRef]
  133. Luckett-Chastain, L.R.; Gallucci, R.M. Interleukin (IL)-6 modulates transforming growth factor-beta expression in skin and dermal fibroblasts from IL-6-deficient mice. Br. J. Dermatol. 2009, 161, 237–248. [Google Scholar] [CrossRef] [PubMed]
  134. Lazarenko, L.M.; Babenko, L.P.; Gichka, S.G.; Sakhno, L.O.; Demchenko, O.M.; Bubnov, R.V.; Sichel, L.M.; Spivak, M.Y. Assessment of the Safety of Lactobacillus casei IMV B-7280 Probiotic Strain on a Mouse Model. Probiotics Antimicrob. Proteins 2021, 13, 1644–1657. [Google Scholar]
  135. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
  136. Bachiller, S.; Ferrer, I.J.; Paulus, A.; Yang, Y.; Swanberg, M.; Deierborg, T.; Boza-Serrano, A. Microglia in Neurological Diseases: A Road Map to Brain-Disease Dependent-Inflammatory Response. Front. Cell. Neurosci. 2018, 12, 488. [Google Scholar] [CrossRef] [PubMed]
  137. Lorente-Picón, M.; Laguna, A. New Avenues for Parkinson’s Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021, 11, 433. [Google Scholar] [CrossRef]
  138. Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics May Have Beneficial Effects in Parkinson’s disease: In vitro Evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef]
  139. Akbari, E.; Asemi, Z.; Kakhaki, R.D.; Bahmani, F.; Kouchaki, E.; Tamtaji, O.R.; Hamidi, G.A.; Salami, M. Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2016, 8, 256. [Google Scholar] [CrossRef]
  140. Agahi, A.; Hamidi, G.A.; Daneshvar, R.; Hamdieh, M.; Soheili, M.; Alinaghipour, A.; Esmaeili Taba, S.M.; Salami, M. Does Severity of Alzheimer’s Disease Contribute to Its Responsiveness to Modifying Gut Microbiota? A Double Blind Clinical Trial. Front. Neurol. 2018, 9, 662. [Google Scholar] [CrossRef]
  141. Tamtaji, O.R.; Heidari-soureshjani, R.; Mirhosseini, N.; Kouchaki, E.; Bahmani, F.; Aghadavod, E.; Tajabadi-Ebrahimi, M.; Asemi, Z. Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in Alzheimer’s disease: A randomized, double-blind, controlled trial. Clin. Nutr. 2019, 38, 2569–2575. [Google Scholar] [CrossRef]
  142. Leblhuber, F.; Steiner, K.; Schuetz, B.; Fuchs, D.; Gostner, J.M. Probiotic Supplementation in Patients with Alzheimer’s De-mentia— An Explorative Intervention Study. Curr. Alzheimer Res. 2018, 15, 1106–1113. [Google Scholar] [CrossRef] [PubMed]
  143. Gerhardt, S.; Mohajeri, M.H. Changes of Colonic Bacterial Composition in Parkinson ’s disease and Other Neurodegenerative Diseases. Nutrients 2018, 10, 708. [Google Scholar] [CrossRef] [PubMed]
  144. Marcos-Fernández, R.; Blanco-Míguez, A.; Ruiz, L.; Margolles, A.; Ruas-Madiedo, P.; Sánchez, B. Towards the isolation of more robust next generation probiotics: The first aerotolerant Bifidobacterium bifidum strain. Food Res. Int. 2023, 165, 112481. [Google Scholar] [CrossRef]
  145. Aponte, M.; Murru, N.; Shoukat, M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front. Microbiol. 2020, 11, 562048. [Google Scholar] [CrossRef] [PubMed]
  146. Salehipour, Z.; Haghmorad, D.; Sankian, M.; Rastin, M.; Nosratabadi, R.; Dallal, M.M.S.; Tabasi, N.; Khazaee, M.; Nasiraii, L.R.; Mahmoudi, M. Bifidobacterium animalis in combination with human origin of Lactobacillus plantarum ameliorate neuroinflammation in experimental model of multiple sclerosis by altering CD4+ T cell subset balance. Biomed. Pharmacother. 2017, 95, 1535–1548. [Google Scholar] [CrossRef]
  147. Kittakoop, P. Contribution of Gut Microbiome to Human Health and the Metabolism or Toxicity of Drugs and Natural Products. In Human Microbiome; IntechOpen: Rijeka, Croatia, 2021. [Google Scholar] [CrossRef]
Figure 1. Identification and screening for literature search.
Figure 1. Identification and screening for literature search.
Microorganisms 11 01083 g001
Figure 2. Microbiota–gut–brain axis. Through neural, metabolic, endocrine, and immunological mechanisms, the brain and stomach interact. The vagus nerve, the hypothalamic-pituitary-adrenal (HPA) axis, and systemic circulation are all influenced by the brain’s function on gastrointestinal health. Short-chain fatty acids (SCFAs), neurotransmitters, and amino acids are examples of signals from the stomach that modify brain activity through neuronal cells, the immune system, and hormonal processes.
Figure 2. Microbiota–gut–brain axis. Through neural, metabolic, endocrine, and immunological mechanisms, the brain and stomach interact. The vagus nerve, the hypothalamic-pituitary-adrenal (HPA) axis, and systemic circulation are all influenced by the brain’s function on gastrointestinal health. Short-chain fatty acids (SCFAs), neurotransmitters, and amino acids are examples of signals from the stomach that modify brain activity through neuronal cells, the immune system, and hormonal processes.
Microorganisms 11 01083 g002
Figure 3. The composition of the microbiome varies with the position of the organ in the human body. This figure shows promising bacterial genera in the stomach, colon, and small intestine.
Figure 3. The composition of the microbiome varies with the position of the organ in the human body. This figure shows promising bacterial genera in the stomach, colon, and small intestine.
Microorganisms 11 01083 g003
Table 1. Inclusion and exclusion criteria.
Table 1. Inclusion and exclusion criteria.
Inclusion CriteriaExclusion Criteria
Study designReview articles and research studies that investigated probiotics and Neurodegenerative diseases Case reports, Hypothesis, Communication, Systematic review and Current opinion
PopulationHumans with no age restriction and any vertebrate or invertebrate model system studiesN/A
Table 3. Study of neurodegenerative diseases and their mediation with probiotics on different invertebrate and vertebrate model systems.
Table 3. Study of neurodegenerative diseases and their mediation with probiotics on different invertebrate and vertebrate model systems.
Model SystemNDMediationOutcomeRefs.
Study History in an Invertebrate Model System
C. elegansPDB. subtilis PXN21
Lactobacillus plantarum
Inhibits and reverses α-syn aggregation,
improved locomotion and reduced dopaminergic neuron degeneration
[108]
ADE. coli OP50
B. subtilis NCIB3610
Alleviated the paralysis phenotype, behavioral deficits, and aggravate lifespan[109]
ALSLacticaseibacillus rhamnosus HA-114Suppresses the progression of motor neuron degeneration[110]
HDBacillus subtilisExtends longevity through downregulation of the insulin-like signaling pathway[111]
D. melanogaster
Park25 flies
ADLactobacillus plantarum DR7Ameliorate the AD effects[112]
Bifidobacterium longum ssp. infantis NCIM 702 255Rescued amyloid beta deposition and acetylcholinesterase activity[113]
Lactobacillus spp.Improved gene expression related to insulin signaling, fat metabolism[114]
PDAcetobacter tropicalisReduced mitochondrial function and disrupted insulin-like signaling and glucose regulation [115,116]
ALSLactobacillus plantarumDegeneration of motor nerve cells[117]
HDE. coliRegulating HTT aggregates and motor defects[118]
Study history in vertebrate model system
3 × Tg AD † miceADLactobacillus plantarum PS128Regulated glycogen synthase kinase 3 beta activity[119]
Bifidobacterium breve A1Reduce the expression of Aβ gene[120]
SLAB51It degrade Aβ plaque through CathepsinD [121]
5× FAD Tg miceL. plantarum C29
Bifidobacterium lactis CUL 34
Decrease the expression of Caspase 3 and increase the activation of microglial[122]
Wistar ratsL. plantarum MTCC 1325Increase the concentration of ACh in hippocampus[123]
Patient (Infant aged < 7 months)B. breve M-16VNo effect on AD marker[124]
Pregnant womanB. longum BB536Prevent eczema and AD in their offspring[125]
Children aged (1–3 years)L. fermentum
L. plantarum CJLP133
Decrease IFNγ and IL4 [126]
Adult patients B. animalis LKM512Reduce the cumulative incidence of AD[127]
MitoPark PD † micePDLactobacillus rhamnosus GG
Bifidobacterium bifidum
Decrease the level of dopaminergic neuronal loss[128]
C57BL/6 mice MPTP † -induced PDStreptococcus thermophilus CRL808Increase pro-inflammatory receptors concentration (IL10)
Bifidobacterium bifidum
Lactobacillus reutri
Lactobacillus fermentum
Increase PPARγ
6-OHDA † induced PD † in C57BL/6 miceSLAB51:
Streptococcus thermophilus,
Bifidobacterium longum,
Bifidobacterium breve,
Bifidobacterium infantis,
Lactobacillus acidophilus,
Lactobacillus plantarum,
Lactobacillus paracasei,
Lactobacillus delbrueckii sub sp. bulgaricus, Lactobacillus brevis
Induces hippocampal long-term
potentiation through BDNF
[129]
PD patientsStreptococcus salivarius sub sp. thermophilus, Enterococcus faecium,
Lactobacillus rhamnosus GG,
Lactobacillus acidophilus
Decrease IL1, IL8 and TNFα[130]
Bifidobacterium infantisReduce abdominal pain and constipation
MiceALSLacticaseibacillus rhamnosus HA-114Through mitochondrial β-oxidation restores lipid equilibrium and energy balance[131]
C57BL/6 mice HDBifidobacterium spp.decreased Il-1β and Il-6 and increased TGF-β and Il-10 expression[132]
BALB/c miceL. casei IMV B-7280Inhibiting NF-κB pathway [133]
C57/BL6 miceL. lactis NZ9000SHD5reduced colonic glandular structure,
downregulated expression of inflammatory molecule
[132]
(† = Disease induced mice model).
Table 4. Recent clinical studies of ND with combination or singular probiotic with their therapeutic effect and mechanism of action.
Table 4. Recent clinical studies of ND with combination or singular probiotic with their therapeutic effect and mechanism of action.
NDProbioticsDurationMechanismTherapeutic EffectRef.
ADL. acidophilus
L. cases
B. bifidum
L. fermentum
12 weeksManagement of metabolic deviationDecreased level of triglyceride in blood
Decreased serum malondialdehyde
Unproductive on antioxidant capacity.
[138]
L. fermentum
L. plantarum
B. lactis
L. acidophilus
B. bifidum
B. longum
12 weeksManagement of Urea and Glucose levelImproved cognitive function.
Unproductive on antioxidant capacity. Improved level of Glutathione
Reduced level of Deoxyguanosine.
[139]
L. acidophilus
B. bifidum
B. longum
selenium
12 weeksMetabolic deviation and oxidative stress balancingDecreased level of triglyceride in blood
Improved the level of Glutathione
Improved antioxidant capacity.
[140]
L. casei W56
L. lactis W19
L. acidophilus W22
B. lactis W52
L. paracasei W20
L. plantarum W62
B. lactis W51
B. bifidum W23
L salivarius W24
28 daysSimulating the microbiota gut brain axis and active immune cellsImproved Faecalibacterium concentration in faecal.
High concentration of kynurenine in blood.
Improved RNA concentration in faecal bacteria.
[141]
PDBlautia
Roseburia,
Coprococcus
Firmicutes
Proteobacteria
Verrucomicrobia, Oscillospira
Bacteroides
NAMaintaining level of fecal microbiota and mucosal levelPro inflammatory dysbiosis,
trigger inflammation
Induced misfolding of α synuclein.
[142]
B. bifidumNARegulation of neuronal inflammationReduced neuroinflammation.[143]
L. salivarius
L. plantarum
L. acidophilus
L. rhamnosus
B. animalis subsp.
B. breve
NA-Monitoring the production of cytokine, superoxide anions (O2-);
Proliferation of E. coli and K. pneumoniae DNA for tyrosine decarboxylase
Reduced ROS generation, improved constipation, and inhibition of E. coli and K. pneumonia all contribute to anti-inflammatory activity.[144]
Multiple SclerosisB. animalisNARegulation of gut barrier permeabilityImproved gut barrier function and reduced inflammation.[145]
ALSL. rhamnosusNARegulation of behavioral changesImproved behavior and social interaction.[146]
B. longumNARegulation of stress and anxiety levelReduced anxiety and depressive symptoms.[147]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ojha, S.; Patil, N.; Jain, M.; Kole, C.; Kaushik, P. Probiotics for Neurodegenerative Diseases: A Systemic Review. Microorganisms 2023, 11, 1083. https://doi.org/10.3390/microorganisms11041083

AMA Style

Ojha S, Patil N, Jain M, Kole C, Kaushik P. Probiotics for Neurodegenerative Diseases: A Systemic Review. Microorganisms. 2023; 11(4):1083. https://doi.org/10.3390/microorganisms11041083

Chicago/Turabian Style

Ojha, Sandhya, Nil Patil, Mukul Jain, Chittaranjan Kole, and Prashant Kaushik. 2023. "Probiotics for Neurodegenerative Diseases: A Systemic Review" Microorganisms 11, no. 4: 1083. https://doi.org/10.3390/microorganisms11041083

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop