Probiotics for Neurodegenerative Diseases: Comparison
Please note this is a comparison between Version 5 by Jessie Wu and Version 4 by Prashant Kaushik.

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 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. 

  • probiotics
  • neurodegenerative diseases
  • gut microbiota
  • aging
  • microglia

1. 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 Alzheimer’s disease (AD), Parkinson’s disease (PD), and others disorders [1]. 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 [2]. 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) [3]. 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 [4]. 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 [5]. 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 [6][7]. 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 [8].

1.1. 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 [9]. 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 [6]. 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 [10].

1.2. 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 [11]. Small intestinal bacterial outgrowth and helicobacter pylori infection has seen in diseased person that causes motor impairment and problem-related with stool [12]. In most cases, patients suffer from increased mucosal permeability and endo-toxic exposure caused by Coliform bacteria [13]. 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 [14]. 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 [15].

1.3. 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 [16][17]. 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 [18][19][20]. 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 [21].

1.4. 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 [22][23]. Bacteria out-growth downregulates lipid and glucose metabolism which disturbs the ATP binding cassette and transporter-associated pathways [24]. 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 [25]. Bulbar function slowly deteriorated with the progression of the disease, and dysphagia (because of aspiration pneumonia and weight loss) have seen [26]. 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 [27]. 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 [28].

2. 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 neurodegenerative disorders (ND) [29]. 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 [30]. 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 [31]. 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 [32]. 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 [33].

2.1. 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 [34]. 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) [35]. In the PD model (C. elegans), expression of α-synuclein was expressed in disease the condition [36]. 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 [37]. The PXN21 protein from B. subtilis prevents and prevents α-synuclein from aggregating [38]. 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 [39]. 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 [40]. 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 [41]. 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 [42]. 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 [43]. In a demethylase-dependent manner, the KDM5 protein controls the immune deficiency (IMD) signaling pathway and maintains bacterial balance in D. melanogaster [44].

2.2. 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 [45]. 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 [46]. 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 [47]. By shedding light on the underlying mechanisms of these conditions and testing the efficacy of probiotics in vertebrate models, researchers might develop treatments that fight against these terrible illnesses Table 1.

3. 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 [74].
Based on the results of both the animal and human studies, the consumption of probiotics has a significant beneficial effect on AD Table 1. WResearchers can conclude that most of the study is based on Bifidobacterium and Lactobacillus and as an outcome this research reveals that probiotics can improve memory dysfunction and cognitive dysfunction in similar to neurodegenerative diseases [38][75]. 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 [76]. 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 2, 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 [77]. 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 [78]. In relation to other ND Lacticaseibacillus rhamnosus HA-114 can improve the energy balance and cholesterol homeostasis in ND animals [71]. In relation to neurodegenerative disease, only 10% of the research concentrates on Streptococcus and Clostridium species. So on, wresearchers give a summary of the literature in this systematic reviewsearch by using data that was extracted from different sources.

References

  1. 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.
  2. 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.
  3. 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.
  4. Rieder, R.; Wisniewski, P.J.; Alderman, B.L.; Campbell, S.C. Microbes and mental health: A review. Brain Behav. Immun. 2017, 66, 9–17.
  5. Nagahara, A.H.; Tuszynski, M.H. Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat. Rev. Drug Discov. 2011, 10, 209–219.
  6. Bell, K.F.; Hardingham, G.E. The influence of synaptic activity on neuronal health. Curr. Opin. Neurobiol. 2011, 21, 299–305.
  7. 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.
  8. 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.
  9. Wu, C.; Sun, D. GABA receptors in brain development, function, and injury. Metab. Brain Dis. 2015, 30, 367–379.
  10. 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.
  11. 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.
  12. 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.
  13. 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.
  14. 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.
  15. 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.
  16. Derkinderen, P.; Shannon, K.M.; Brundin, P. Gut feelings about smoking and coffee in Parkinson’s disease. Mov. Disord. 2014, 29, 976–979.
  17. 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.
  18. 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.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. Round, J.L.; Mazmanian, S.K. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 2009, 9, 313–323.
  26. 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.
  27. 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.
  28. Rowin, J.; Xia, Y.; Jung, B.; Sun, J. Gut inflammation and dysbiosis in human motor neuron disease. Physiol. Rep. 2017, 5, e13443.
  29. 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.
  30. 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.
  31. Pajares, M.; IRojo, A.; Manda, G.; Boscá, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells 2020, 9, 1687.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272.
  37. 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.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. Salles, B.I.M.; Cioffi, D.; Ferreira, S.R.G. Probiotics supplementation and insulin resistance: A systematic review. Diabetol. Metab. Syndr. 2020, 12, 98.
  57. 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.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. Wong, C.B.; Iwabuchi, N.; Xiao, J.-Z. Exploring the Science behind Bifidobacterium breve M-16V in Infant Health. Nutrients 2019, 11, 1724.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. Lorente-Picón, M.; Laguna, A. New Avenues for Parkinson’s Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021, 11, 433.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. Gerhardt, S.; Mohajeri, M.H. Changes of Colonic Bacterial Composition in Parkinson ’s disease and Other Neurodegenerative Diseases. Nutrients 2018, 10, 708.
  85. 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.
  86. Aponte, M.; Murru, N.; Shoukat, M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front. Microbiol. 2020, 11, 562048.
  87. 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.
  88. 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.
More
Video Production Service