Microecologics and Exercise: Targeting the Microbiota–Gut–Brain Axis for Central Nervous System Disease Intervention
Abstract
:1. Introduction
2. Central Nervous System Diseases and Microbiome–Gut–Brain Axis
2.1. Central Nervous System Diseases
2.1.1. Alzheimer’s Disease
2.1.2. Parkinson’s Disease
2.1.3. Autism Spectrum Disorder
2.1.4. Multiple Sclerosis
2.2. Microbiota–Gut–Brain Axis
2.2.1. Immune Pathways
2.2.2. Endocrine Pathway
2.2.3. Neuronal Pathway
3. The Impact of Microecologics on Central Nervous System Diseases
3.1. Classification and Functions of Microecologics
3.1.1. Probiotics
3.1.2. Prebiotics
3.1.3. Synbiotics
3.1.4. Postbiotics
3.2. Mechanisms by Which Microecologics Exert Effects on Central Nervous System Diseases
3.2.1. Enhancement of Intestinal Barrier Function
3.2.2. Regulation of the Blood–Brain Barrier
3.2.3. Regulation of Intestinal Immune Function
3.2.4. Regulation of Neurotransmitters and the Vagus Nerve
3.2.5. Regulation of Gut Microbiota
4. The Influence of Exercise on Central Nervous System Diseases
4.1. Modulatory Effects of Exercise on Central Nervous System Diseases
4.2. Possible Mechanisms by Which Exercise Exerts Its Effects on Central Nervous System Diseases
4.2.1. The Anti-Neuroinflammatory and Immunomodulatory Effects of Exercise
4.2.2. The Antioxidant Effect of Exercise
4.2.3. Exercise Regulates Neurotransmitters and Neural Plasticity
4.2.4. Exercise Regulates Neurotrophic Factors
4.2.5. Exercise Modulates Gut Microbiota
5. The Possibility of Combined Intervention of Microecologics and Exercise on Central Nervous System Diseases
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
References
- Ji, N.; Wang, F.; Wang, M.; Zhang, W.; Liu, H.; Su, J. Engineered bacterial extracellular vesicles for central nervous system diseases. J. Control Release 2023, 364, 46–60. [Google Scholar] [CrossRef] [PubMed]
- Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
- Agnello, L.; Ciaccio, M. Neurodegenerative Diseases: From Molecular Basis to Therapy. Int. J. Mol. Sci. 2022, 23, 12854. [Google Scholar] [CrossRef]
- Heemels, M.-T. Neurodegenerative diseases. Nature 2016, 539, 179. [Google Scholar] [CrossRef]
- Checkoway, H.; Lundin, J.I.; Kelada, S.N. Neurodegenerative diseases. IARC Sci. Publ. 2011, 163, 407–419. [Google Scholar]
- Voon, V.; Napier, T.C.; Frank, M.J.; Sgambato-Faure, V.; Grace, A.A.; Rodriguez-Oroz, M.; Obeso, J.; Bezard, E.; Fernagut, P.-O. Impulse control disorders and levodopa-induced dyskinesias in Parkinson’s disease: An update. Lancet Neurol. 2017, 16, 238–250. [Google Scholar] [CrossRef]
- Mak, M.K.; Wong-Yu, I.S.; Shen, X.; Chung, C.L. Long-term effects of exercise and physical therapy in people with Parkinson disease. Nat. Rev. Neurol. 2017, 13, 689–703. [Google Scholar] [CrossRef]
- Yang, K.; Chen, Y.; Wang, M.; Zhang, Y.; Yuan, Y.; Hou, H.; Mao, Y.-H. The Improvement and Related Mechanism of Microecologics on the Sports Performance and Post-Exercise Recovery of Athletes: A Narrative Review. Nutrients 2024, 16, 1602. [Google Scholar] [CrossRef]
- Sujkowski, A.; Hong, L.; Wessells, R.J.; Todi, S.V. The protective role of exercise against age-related neurodegeneration. Ageing Res. Rev. 2022, 74, 101543. [Google Scholar] [CrossRef]
- Liu, Q.; Xi, Y.; Wang, Q.; Liu, J.; Li, P.; Meng, X.; Liu, K.; Chen, W.; Liu, X.; Liu, Z. Mannan oligosaccharide attenuates cognitive and behavioral disorders in the 5xFAD Alzheimer’s disease mouse model via regulating the gut microbiota-brain axis. Brain Behav. Immun. 2021, 95, 330–343. [Google Scholar] [CrossRef] [PubMed]
- Valenzuela, P.L.; Castillo-García, A.; Morales, J.S.; de la Villa, P.; Hampel, H.; Emanuele, E.; Lista, S.; Lucia, A. Exercise benefits on Alzheimer’s disease: State-of-the-science. Ageing Res. Rev. 2020, 62, 101108. [Google Scholar] [CrossRef] [PubMed]
- Ansari, F.; Pourjafar, H.; Tabrizi, A.; Homayouni, A. The Effects of Probiotics and Prebiotics on Mental Disorders: A Review on Depression, Anxiety, Alzheimer, and Autism Spectrum Disorders. Curr. Pharm. Biotechnol. 2020, 21, 555–565. [Google Scholar] [CrossRef]
- Heine, M.; van de Port, I.; Rietberg, M.B.; van Wegen, E.E.H.; Kwakkel, G. Exercise therapy for fatigue in multiple sclerosis. Cochrane Database Syst. Rev. 2015, 2015, CD009956. [Google Scholar] [CrossRef]
- Kola, S.; Subramanian, I. Updates in Parkinson’s Disease Integrative Therapies: An Evidence-Based Review. Curr. Neurol. Neurosci. Rep. 2023, 23, 717–726. [Google Scholar] [CrossRef]
- Abraham, D.; Feher, J.; Scuderi, G.L.; Szabo, D.; Dobolyi, A.; Cservenak, M.; Juhasz, J.; Ligeti, B.; Pongor, S.; Gomez-Cabrera, M.C.; et al. Exercise and probiotics attenuate the development of Alzheimer’s disease in transgenic mice: Role of microbiome. Exp. Gerontol. 2019, 115, 122–131. [Google Scholar] [CrossRef]
- Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.-F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflamm. 2019, 16, 53. [Google Scholar] [CrossRef]
- Teng, M.; Zhao, X.; Wang, C.; Wang, C.; White, J.C.; Zhao, W.; Zhou, L.; Duan, M.; Wu, F. Polystyrene Nanoplastics Toxicity to Zebrafish: Dysregulation of the Brain-Intestine-Microbiota Axis. ACS Nano 2022, 16, 8190–8204. [Google Scholar] [CrossRef]
- Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
- Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef]
- Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef]
- Liu, P.; Wu, L.; Peng, G.; Han, Y.; Tang, R.; Ge, J.; Zhang, L.; Jia, L.; Yue, S.; Zhou, K.; et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav. Immun. 2019, 80, 633–643. [Google Scholar] [CrossRef] [PubMed]
- Zha, X.; Liu, X.; Wei, M.; Huang, H.; Cao, J.; Liu, S.; Bian, X.; Zhang, Y.; Xiao, F.; Xie, Y.; et al. Microbiota-derived lysophosphatidylcholine alleviates Alzheimer’s disease pathology via suppressing ferroptosis. Cell Metabo. 2025, 37, 169–186.e169. [Google Scholar] [CrossRef] [PubMed]
- Harach, T.; Marungruang, N.; Duthilleul, N.; Cheatham, V.; Mc Coy, K.D.; Frisoni, G.; Neher, J.J.; Fåk, F.; Jucker, M.; Lasser, T.; et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci. Rep. 2017, 7, 41802. [Google Scholar] [CrossRef]
- Scheperjans, F.; Aho, V.; Pereira, P.A.B.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 1351–1360. [Google Scholar] [CrossRef]
- Sun, M.F.; Zhu, Y.L.; Zhou, Z.L.; Jia, X.B.; Xu, Y.D.; Yang, Q.; Cui, C.; Shen, Y.Q. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav. Immun. 2018, 70, 48–60. [Google Scholar] [CrossRef]
- Gorecki, A.M.; Preskey, L.; Bakeberg, M.C.; Kenna, J.E.; Gildenhuys, C.; MacDougall, G.; Dunlop, S.A.; Mastaglia, F.L.; Akkari, P.A.; Koengten, F.; et al. Altered Gut Microbiome in Parkinson’s Disease and the Influence of Lipopolysaccharide in a Human α-Synuclein Over-Expressing Mouse Model. Front. Neurosci. 2019, 13, 839. [Google Scholar] [CrossRef]
- Pulikkan, J.; Maji, A.; Dhakan, D.B.; Saxena, R.; Mohan, B.; Anto, M.M.; Agarwal, N.; Grace, T.; Sharma, V.K. Gut Microbial Dysbiosis in Indian Children with Autism Spectrum Disorders. Microb. Ecol. 2018, 76, 1102–1114. [Google Scholar] [CrossRef]
- Sharon, G.; Cruz, N.J.; Kang, D.-W.; Gandal, M.J.; Wang, B.; Kim, Y.-M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; Lane, C.J.; et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019, 177, 1600–1618.e17. [Google Scholar] [CrossRef]
- Chen, J.; Chia, N.; Kalari, K.R.; Yao, J.Z.; Novotna, M.; Paz Soldan, M.M.; Luckey, D.H.; Marietta, E.V.; Jeraldo, P.R.; Chen, X.; et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci. Rep. 2016, 6, 28484. [Google Scholar] [CrossRef]
- Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed]
- Feng, R.; Zhu, Q.; Wang, A.; Wang, H.; Wang, J.; Chen, P.; Zhang, R.; Liang, D.; Teng, J.; Ma, M.; et al. Effect of fecal microbiota transplantation on patients with sporadic amyotrophic lateral sclerosis: A randomized, double-blind, placebo-controlled trial. BMC Med. 2024, 22, 566. [Google Scholar] [CrossRef]
- Mazzini, L.; Mogna, L.; De Marchi, F.; Amoruso, A.; Pane, M.; Aloisio, I.; Cionci, N.B.; Gaggìa, F.; Lucenti, A.; Bersano, E.; et al. Potential Role of Gut Microbiota in ALS Pathogenesis and Possible Novel Therapeutic Strategies. J. Clin. Gastroenterol. 2018, 52 (Suppl. 1), S68–S70. [Google Scholar] [CrossRef]
- Wasser, C.I.; Mercieca, E.C.; Kong, G.; Hannan, A.J.; McKeown, S.J.; Glikmann-Johnston, Y.; Stout, J.C. Gut dysbiosis in Huntington’s disease: Associations among gut microbiota, cognitive performance and clinical outcomes. Brain Commun. 2020, 2, fcaa110. [Google Scholar] [CrossRef]
- Fang, P.; Kazmi, S.A.; Jameson, K.G.; Hsiao, E.Y. The Microbiome as a Modifier of Neurodegenerative Disease Risk. Cell Host Microbe 2020, 28, 201–222. [Google Scholar] [CrossRef]
- Kong, G.; Cao, K.L.; Judd, L.M.; Li, S.; Renoir, T.; Hannan, A.J. Microbiome profiling reveals gut dysbiosis in a transgenic mouse model of Huntington’s disease. Neurobiol. Dis. 2020, 135, 104268. [Google Scholar] [CrossRef]
- Wang, L.J.; Yang, C.Y.; Kuo, H.C.; Chou, W.J.; Tsai, C.S.; Lee, S.Y. Effect of Bifidobacterium bifidum on Clinical Characteristics and Gut Microbiota in Attention-Deficit/Hyperactivity Disorder. J. Pers. Med. 2022, 12, 227. [Google Scholar] [CrossRef]
- Wang, L.J.; Yang, C.Y.; Chou, W.J.; Lee, M.J.; Chou, M.C.; Kuo, H.C.; Yeh, Y.M.; Lee, S.Y.; Huang, L.H.; Li, S.C. Gut microbiota and dietary patterns in children with attention-deficit/hyperactivity disorder. Eur. Child Adolesc. Psychiatry 2020, 29, 287–297. [Google Scholar] [CrossRef]
- Twarowski, B.; Herbet, M. Inflammatory Processes in Alzheimer’s Disease-Pathomechanism, Diagnosis and Treatment: A Review. Int. J. Mol. Sci. 2023, 24, 6518. [Google Scholar] [CrossRef] [PubMed]
- Soscia, S.J.; Kirby, J.E.; Washicosky, K.J.; Tucker, S.M.; Ingelsson, M.; Hyman, B.; Burton, M.A.; Goldstein, L.E.; Duong, S.; Tanzi, R.E.; et al. The Alzheimer’s disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS ONE 2010, 5, e9505. [Google Scholar] [CrossRef]
- Morris, G.; Berk, M.; Maes, M.; Puri, B.K. Could Alzheimer’s Disease Originate in the Periphery and If So How So? Mol. Neurobiol. 2019, 56, 406–434. [Google Scholar] [CrossRef] [PubMed]
- Mancuso, C.; Santangelo, R. Alzheimer’s disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol. Res. 2018, 129, 329–336. [Google Scholar] [CrossRef]
- Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
- Chen, C.; Liao, J.; Xia, Y.; Liu, X.; Jones, R.; Haran, J.; McCormick, B.; Sampson, T.R.; Alam, A.; Ye, K. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut 2022, 71, 2233–2252. [Google Scholar] [CrossRef]
- Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Grigoriadis, N.; Deretzi, G.; Tzilves, D.; Katsinelos, P.; Tsolaki, M.; Chatzopoulos, D.; et al. Eradication of Helicobacter pylori may be beneficial in the management of Alzheimer’s disease. J. Neurol. 2009, 256, 758–767. [Google Scholar] [CrossRef]
- Hayes, M.T. Parkinson’s Disease and Parkinsonism. Am. J. Med. 2019, 132, 802–807. [Google Scholar] [CrossRef]
- Schneider, S.A.; Alcalay, R.N. Neuropathology of genetic synucleinopathies with parkinsonism: Review of the literature. Mov. Disord. 2017, 32, 1504–1523. [Google Scholar] [CrossRef]
- Lebouvier, T.; Chaumette, T.; Paillusson, S.; Duyckaerts, C.; Bruley des Varannes, S.; Neunlist, M.; Derkinderen, P. The second brain and Parkinson’s disease. Eur. J. Neurosci. 2009, 30, 735–741. [Google Scholar] [CrossRef]
- Natale, G.; Pasquali, L.; Paparelli, A.; Fornai, F. Parallel manifestations of neuropathologies in the enteric and central nervous systems. Neurogastroenterol. Motil. 2011, 23, 1056–1065. [Google Scholar] [CrossRef]
- Cannon, T.; Gruenheid, S. Microbes and Parkinson’s disease: From associations to mechanisms. Trends Microbiol. 2022, 30, 749–760. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [PubMed]
- Bhattarai, Y.; Si, J.; Pu, M.; Ross, O.A.; McLean, P.J.; Till, L.; Moor, W.; Grover, M.; Kandimalla, K.K.; Margolis, K.G.; et al. Role of gut microbiota in regulating gastrointestinal dysfunction and motor symptoms in a mouse model of Parkinson’s disease. Gut Microbes 2021, 13, 1866974. [Google Scholar] [CrossRef]
- Loh, J.S.; Mak, W.Q.; Tan, L.K.S.; Ng, C.X.; Chan, H.H.; Yeow, S.H.; Foo, J.B.; Ong, Y.S.; How, C.W.; Khaw, K.Y. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct. Target. Ther. 2024, 9, 37. [Google Scholar]
- Wang, Q.; Yang, Q.; Liu, X. The microbiota-gut-brain axis and neurodevelopmental disorders. Protein Cell 2023, 14, 762–775. [Google Scholar] [CrossRef]
- Oliver, L.D.; Moxon-Emre, I.; Lai, M.-C.; Grennan, L.; Voineskos, A.N.; Ameis, S.H. Social Cognitive Performance in Schizophrenia Spectrum Disorders Compared With Autism Spectrum Disorder: A Systematic Review, Meta-analysis, and Meta-regression. JAMA Psychiatry 2021, 78, 281–292. [Google Scholar] [CrossRef]
- Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism spectrum disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef]
- Li, Z.; Zhu, Y.-X.; Gu, L.-J.; Cheng, Y. Understanding autism spectrum disorders with animal models: Applications, insights, and perspectives. Zool. Res. 2021, 42, 800–824. [Google Scholar] [CrossRef]
- McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal symptoms in autism spectrum disorder: A meta-analysis. Pediatrics 2014, 133, 872–883. [Google Scholar] [CrossRef]
- Strati, F.; Cavalieri, D.; Albanese, D.; De Felice, C.; Donati, C.; Hayek, J.; Jousson, O.; Leoncini, S.; Renzi, D.; Calabrò, A.; et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome 2017, 5, 24. [Google Scholar] [CrossRef]
- Kang, D.-W.; Park, J.G.; Ilhan, Z.E.; Wallstrom, G.; Labaer, J.; Adams, J.B.; Krajmalnik-Brown, R. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS ONE 2013, 8, e68322. [Google Scholar] [CrossRef]
- Liu, F.; Horton-Sparks, K.; Hull, V.; Li, R.W.; Martínez-Cerdeño, V. The valproic acid rat model of autism presents with gut bacterial dysbiosis similar to that in human autism. Mol. Autism 2018, 9, 61. [Google Scholar] [CrossRef] [PubMed]
- Emanuele, E.; Orsi, P.; Boso, M.; Broglia, D.; Brondino, N.; Barale, F.; di Nemi, S.U.; Politi, P. Low-grade endotoxemia in patients with severe autism. Neurosci. Lett. 2010, 471, 162–165. [Google Scholar] [CrossRef]
- Kang, D.-W.; Adams, J.B.; Gregory, A.C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S.; et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study. Microbiome 2017, 5, 10. [Google Scholar] [CrossRef]
- Porcari, S.; Benech, N.; Valles-Colomer, M.; Segata, N.; Gasbarrini, A.; Cammarota, G.; Sokol, H.; Ianiro, G. Key determinants of success in fecal microbiota transplantation: From microbiome to clinic. Cell Host Microbe 2023, 31, 712–733. [Google Scholar] [CrossRef]
- Yu, Y.; Zhao, F. Microbiota-gut-brain axis in autism spectrum disorder. J. Genet. Genom. 2021, 48, 755–762. [Google Scholar] [CrossRef]
- Hemmer, B.; Archelos, J.J.; Hartung, H.-P. New concepts in the immunopathogenesis of multiple sclerosis. Nat. Rev. Neurosci. 2002, 3, 291–301. [Google Scholar] [CrossRef]
- Ramagopalan, S.V.; Dobson, R.; Meier, U.C.; Giovannoni, G. Multiple sclerosis: Risk factors, prodromes, and potential causal pathways. Lancet Neurol. 2010, 9, 727–739. [Google Scholar] [CrossRef]
- Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [Google Scholar] [CrossRef]
- Fletcher, J.M.; Lalor, S.J.; Sweeney, C.M.; Tubridy, N.; Mills, K.H.G. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin. Exp. Immunol. 2010, 162, 1–11. [Google Scholar] [CrossRef]
- Kasper, L.H.; Shoemaker, J. Multiple sclerosis immunology: The healthy immune system vs the MS immune system. Neurology 2010, 74 (Suppl. 1), S2–S8. [Google Scholar] [CrossRef]
- Venken, K.; Hellings, N.; Hensen, K.; Rummens, J.-L.; Medaer, R.; D’Hooghe, M.B.; Dubois, B.; Raus, J.; Stinissen, P. Secondary progressive in contrast to relapsing-remitting multiple sclerosis patients show a normal CD4+CD25+ regulatory T-cell function and FOXP3 expression. J. Neurosci. Res. 2006, 83, 1432–1446. [Google Scholar] [CrossRef] [PubMed]
- Viglietta, V.; Baecher-Allan, C.; Weiner, H.L.; Hafler, D.A. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J. Exp. Med. 2004, 199, 971–979. [Google Scholar] [CrossRef] [PubMed]
- Hohlfeld, R. Multiple sclerosis: Human model for EAE? Eur. J. Immunol. 2009, 39, 2036–2039. [Google Scholar] [CrossRef]
- Berer, K.; Mues, M.; Koutrolos, M.; Rasbi, Z.A.; Boziki, M.; Johner, C.; Wekerle, H.; Krishnamoorthy, G. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 2011, 479, 538–541. [Google Scholar] [CrossRef]
- Croxford, A.L.; Kurschus, F.C.; Waisman, A. Mouse models for multiple sclerosis: Historical facts and future implications. Biochim. Biophys. Acta 2011, 1812, 177–183. [Google Scholar] [CrossRef]
- Ochoa-Repáraz, J.; Mielcarz, D.W.; Ditrio, L.E.; Burroughs, A.R.; Foureau, D.M.; Haque-Begum, S.; Kasper, L.H. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J. Immunol. 2009, 183, 6041–6050. [Google Scholar] [CrossRef] [PubMed]
- Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef]
- Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef]
- Tremlett, H.; Fadrosh, D.W.; Faruqi, A.A.; Hart, J.; Roalstad, S.; Graves, J.; Lynch, S.; Waubant, E. Gut microbiota composition and relapse risk in pediatric MS: A pilot study. J. Neurol. Sci. 2016, 363, 153–157. [Google Scholar] [CrossRef]
- Humphrey, J.; Venkatesh, S.; Hasan, R.; Herb, J.T.; de Paiva Lopes, K.; Küçükali, F.; Byrska-Bishop, M.; Evani, U.S.; Narzisi, G.; Fagegaltier, D.; et al. Integrative transcriptomic analysis of the amyotrophic lateral sclerosis spinal cord implicates glial activation and suggests new risk genes. Nat. Neurosci. 2023, 26, 150–162. [Google Scholar] [CrossRef]
- Tabrizi, S.J.; Estevez-Fraga, C.; van Roon-Mom, W.M.C.; Flower, M.D.; Scahill, R.I.; Wild, E.J.; Muñoz-Sanjuan, I.; Sampaio, C.; Rosser, A.E.; Leavitt, B.R. Potential disease-modifying therapies for Huntington’s disease: Lessons learned and future opportunities. Lancet Neurol. 2022, 21, 645–658. [Google Scholar] [CrossRef] [PubMed]
- You, M.; Chen, N.; Yang, Y.; Cheng, L.; He, H.; Cai, Y.; Liu, Y.; Liu, H.; Hong, G. The gut microbiota-brain axis in neurological disorders. MedComm 2024, 5, e656. [Google Scholar] [CrossRef] [PubMed]
- Aparici-Carratalá, D.; Esclapez, J.; Bautista, V.; Bonete, M.-J.; Camacho, M. Archaea: Current and potential biotechnological applications. Res. Microbiol. 2023, 174, 104080. [Google Scholar] [CrossRef]
- Zhuang, M.; Zhang, X.; Cai, J. Microbiota-gut-brain axis: Interplay between microbiota, barrier function and lymphatic system. Gut Microbes 2024, 16, 2387800. [Google Scholar] [CrossRef]
- Ding, M.; Lang, Y.; Shu, H.; Shao, J.; Cui, L. Microbiota-Gut-Brain Axis and Epilepsy: A Review on Mechanisms and Potential Therapeutics. Front. Immunol. 2021, 12, 742449. [Google Scholar] [CrossRef]
- Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The Brain-Gut-Microbiome Axis. Cell Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef]
- Borkent, J.; Ioannou, M.; Laman, J.D.; Haarman, B.C.M.; Sommer, I.E.C. Role of the gut microbiome in three major psychiatric disorders. Psychol. Med. 2022, 52, 1222–1242. [Google Scholar] [CrossRef]
- Hillestad, E.M.R.; van der Meeren, A.; Nagaraja, B.H.; Bjørsvik, B.R.; Haleem, N.; Benitez-Paez, A.; Sanz, Y.; Hausken, T.; Lied, G.A.; Lundervold, A.; et al. Gut bless you: The microbiota-gut-brain axis in irritable bowel syndrome. World J. Gastroenterol. 2022, 28, 412–431. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Agirman, G.; Hsiao, E.Y. SnapShot: The microbiota-gut-brain axis. Cell 2021, 184, 2524–2524.e1. [Google Scholar] [CrossRef]
- Needham, B.D.; Funabashi, M.; Adame, M.D.; Wang, Z.; Boktor, J.C.; Haney, J.; Wu, W.-L.; Rabut, C.; Ladinsky, M.S.; Hwang, S.-J.; et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647–653. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
- Brescia, P.; Rescigno, M. The gut vascular barrier: A new player in the gut-liver-brain axis. Trends Mol. Med. 2021, 27, 844–855. [Google Scholar] [CrossRef]
- Pellegrini, C.; Antonioli, L.; Colucci, R.; Blandizzi, C.; Fornai, M. Interplay among gut microbiota, intestinal mucosal barrier and enteric neuro-immune system: A common path to neurodegenerative diseases? Acta Neuropathol. 2018, 136, 345–361. [Google Scholar] [CrossRef]
- Wong, J.M.W.; de Souza, R.; Kendall, C.W.C.; Emam, A.; Jenkins, D.J.A. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
- Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain Fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277–288. [Google Scholar] [CrossRef]
- Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of immune cell function by short-chain fatty acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef]
- Malech, H.L.; DeLeo, F.R.; Quinn, M.T. The Role of Neutrophils in the Immune System: An Overview. Methods Mol. Biol. 2020, 2087, 3–10. [Google Scholar]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
- Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome-microglia connections via the gut-brain axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef]
- Gaillard, R.C. Cytokines in the neuroendocrine system. Int. Rev. Immunol. 1998, 17, 181–216. [Google Scholar] [CrossRef] [PubMed]
- Daneman, R.; Prat, A. The blood-brain barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef]
- 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]
- Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef]
- Cussotto, S.; Sandhu, K.V.; Dinan, T.G.; Cryan, J.F. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Front. Neuroendocrinol. 2018, 51, 80–101. [Google Scholar] [CrossRef]
- Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef]
- Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef]
- Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef]
- Isacson, R.; Nielsen, E.; Dannaeus, K.; Bertilsson, G.; Patrone, C.; Zachrisson, O.; Wikström, L. The glucagon-like peptide 1 receptor agonist exendin-4 improves reference memory performance and decreases immobility in the forced swim test. Eur. J. Pharmacol. 2011, 650, 249–255. [Google Scholar] [CrossRef]
- McClean, P.L.; Parthsarathy, V.; Faivre, E.; Hölscher, C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J. Neurosci. 2011, 31, 6587–6594. [Google Scholar] [CrossRef]
- Porter, D.W.; Irwin, N.; Flatt, P.R.; Hölscher, C.; Gault, V.A. Prolonged GIP receptor activation improves cognitive function, hippocampal synaptic plasticity and glucose homeostasis in high-fat fed mice. Eur. J. Pharmacol. 2011, 650, 688–693. [Google Scholar] [CrossRef] [PubMed]
- Lutz-Bucher, B.; González de Aguilar, J.L.; René, F.; Sée, V.; Gordon, J.W.; Loeffler, J. Oxidative stress and a murine superoxide dismutase-1 mutation promoting amyotrophic lateral sclerosis alter neurosecretion in the hypothalamo-neurohypophyseal axis. Neuroendocrinology 1999, 69, 377–384. [Google Scholar] [CrossRef] [PubMed]
- Miaskowski, C.; Ong, G.L.; Lukic, D.; Haldar, J. Immobilization stress affects oxytocin and vasopressin levels in hypothalamic and extrahypothalamic sites. Brain Res. 1988, 458, 137–141. [Google Scholar] [CrossRef]
- Sharan, P.; Vellapandian, C. Hypothalamic-Pituitary-Adrenal (HPA) Axis: Unveiling the Potential Mechanisms Involved in Stress-Induced Alzheimer’s Disease and Depression. Cureus 2024, 16, e67595. [Google Scholar]
- van Dalfsen, J.H.; Markus, C.R. The influence of sleep on human hypothalamic-pituitary-adrenal (HPA) axis reactivity: A systematic review. Sleep. Med. Rev. 2018, 39, 187–194. [Google Scholar] [CrossRef]
- Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259.e6. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.; Zou, J.; Yang, L.; Zhao, J.; Wang, L.; Liu, T.; Fan, X. Alteration of peripheral cortisol and autism spectrum disorder: A meta-analysis. Front. Psychiatry 2022, 13, 928188. [Google Scholar] [CrossRef]
- Wu, W.-L.; Adame, M.D.; Liou, C.-W.; Barlow, J.T.; Lai, T.-T.; Sharon, G.; Schretter, C.E.; Needham, B.D.; Wang, M.I.; Tang, W.; et al. Microbiota regulate social behaviour via stress response neurons in the brain. Nature 2021, 595, 409–414. [Google Scholar] [CrossRef]
- Uesaka, T.; Young, H.M.; Pachnis, V.; Enomoto, H. Development of the intrinsic and extrinsic innervation of the gut. Dev. Biol. 2016, 417, 158–167. [Google Scholar] [CrossRef]
- Marklund, U. Diversity, development and immunoregulation of enteric neurons. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 85–86. [Google Scholar] [CrossRef]
- Yoo, B.B.; Mazmanian, S.K. The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 2017, 46, 910–926. [Google Scholar] [CrossRef] [PubMed]
- Fülling, C.; Dinan, T.G.; Cryan, J.F. Gut Microbe to Brain Signaling: What Happens in Vagus…. Neuron 2019, 101, 998–1002. [Google Scholar] [CrossRef] [PubMed]
- Waise, T.M.Z.; Dranse, H.J.; Lam, T.K.T. The metabolic role of vagal afferent innervation. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 625–636. [Google Scholar] [CrossRef]
- de Lartigue, G.; de La Serre, C.B.; Raybould, H.E. Vagal afferent neurons in high fat diet-induced obesity; intestinal microflora, gut inflammation and cholecystokinin. Physiol. Behav. 2011, 105, 100–105. [Google Scholar] [CrossRef]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
- Bercik, P.; Denou, E.; Collins, J.; Jackson, W.; Lu, J.; Jury, J.; Deng, Y.; Blennerhassett, P.; Macri, J.; McCoy, K.D.; et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 2011, 141, 599–609.e6093. [Google Scholar] [CrossRef]
- Rao, M.; Gershon, M.D. Enteric nervous system development: What could possibly go wrong? Nat. Rev. Neurosci. 2018, 19, 552–565. [Google Scholar] [CrossRef]
- Brun, P.; Giron, M.C.; Qesari, M.; Porzionato, A.; Caputi, V.; Zoppellaro, C.; Banzato, S.; Grillo, A.R.; Spagnol, L.; De Caro, R.; et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 2013, 145, 1323–1333. [Google Scholar] [CrossRef]
- Mao, Y.-K.; Kasper, D.L.; Wang, B.; Forsythe, P.; Bienenstock, J.; Kunze, W.A. Bacteroides fragilis polysaccharide A is necessary and sufficient for acute activation of intestinal sensory neurons. Nat. Commun. 2013, 4, 1465. [Google Scholar] [CrossRef]
- Fröhlich, E.E.; Farzi, A.; Mayerhofer, R.; Reichmann, F.; Jačan, A.; Wagner, B.; Zinser, E.; Bordag, N.; Magnes, C.; Fröhlich, E.; et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav. Immun. 2016, 56, 140–155. [Google Scholar] [CrossRef]
- Wang, M.; Yuan, Y.; Song, A.; Weng, X.; Mao, Y. Research progress in the regulation of gut microbiome by microbioecologics with heavy-load exercise. Life Sci. 2023, 35, 1639–1651. [Google Scholar] [CrossRef]
- Liu, Y.-W.; Liong, M.T.; Chung, Y.-C.E.; Huang, H.-Y.; Peng, W.-S.; Cheng, Y.-F.; Lin, Y.-S.; Wu, Y.-Y.; Tsai, Y.-C. Effects of Lactobacillus plantarum PS128 on Children with Autism Spectrum Disorder in Taiwan: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2019, 11, 820. [Google Scholar] [CrossRef]
- Mehrabadi, S.; Sadr, S.S. Assessment of Probiotics Mixture on Memory Function, Inflammation Markers, and Oxidative Stress in an Alzheimer’s Disease Model of Rats. Iran. Biomed. J. 2020, 24, 220–228. [Google Scholar] [CrossRef]
- Tankou, S.K.; Regev, K.; Healy, B.C.; Tjon, E.; Laghi, L.; Cox, L.M.; Kivisäkk, P.; Pierre, I.V.; Hrishikesh, L.; Gandhi, R.; et al. A probiotic modulates the microbiome and immunity in multiple sclerosis. Ann. Neurol. 2018, 83, 1147–1161. [Google Scholar] [CrossRef]
- Cassani, E.; Privitera, G.; Pezzoli, G.; Pusani, C.; Madio, C.; Iorio, L.; Barichella, M. Use of probiotics for the treatment of constipation in Parkinson’s disease patients. Minerva Gastroenterol. Dietol. 2011, 57, 117–121. [Google Scholar]
- Tamtaji, O.R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1031–1035. [Google Scholar] [CrossRef]
- 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]
- Allahyari, P.; Abbas Torki, S.; Aminnezhad Kavkani, B.; Mahmoudi, Z.; Mousavi Hoseini, M.S.; Moradi, M.; Alami, F.; Keshavarz Mohammadian, M.; Bahoo Sele Bani, S.; Abbasi Mobarakeh, K.; et al. A systematic review of the beneficial effects of prebiotics, probiotics, and synbiotics on ADHD. Neuropsychopharmacol. Rep. 2024, 44, 300–307. [Google Scholar] [CrossRef]
- Blacher, E.; Bashiardes, S.; Shapiro, H.; Rothschild, D.; Mor, U.; Dori-Bachash, M.; Kleimeyer, C.; Moresi, C.; Harnik, Y.; Zur, M.; et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 2019, 572, 474–480. [Google Scholar] [CrossRef]
- Lee, Y.-S.; Lai, D.-M.; Huang, H.-J.; Lee-Chen, G.-J.; Chang, C.-H.; Hsieh-Li, H.M.; Lee, G.-C. Prebiotic Lactulose Ameliorates the Cognitive Deficit in Alzheimer’s Disease Mouse Model through Macroautophagy and Chaperone-Mediated Autophagy Pathways. J. Agric. Food Chem. 2021, 69, 2422–2437. [Google Scholar] [CrossRef]
- Chen, D.; Yang, X.; Yang, J.; Lai, G.; Yong, T.; Tang, X.; Shuai, O.; Zhou, G.; Xie, Y.; Wu, Q. Prebiotic Effect of Fructooligosaccharides from Morinda officinalis on Alzheimer’s Disease in Rodent Models by Targeting the Microbiota-Gut-Brain Axis. Front. Aging Neurosci. 2017, 9, 403. [Google Scholar] [CrossRef] [PubMed]
- Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejía, J.L.; Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome 2018, 6, 133. [Google Scholar] [CrossRef]
- Liu, X.; Du, Z.R.; Wang, X.; Sun, X.R.; Zhao, Q.; Zhao, F.; Wong, W.T.; Wong, K.H.; Dong, X.-L. Polymannuronic acid prebiotic plus Lacticaseibacillus rhamnosus GG probiotic as a novel synbiotic promoted their separate neuroprotection against Parkinson’s disease. Food Res. Int. 2022, 155, 111067. [Google Scholar] [CrossRef]
- Liu, J.; Wang, F.; Liu, S.; Du, J.; Hu, X.; Xiong, J.; Fang, R.; Chen, W.; Sun, J. Sodium butyrate exerts protective effect against Parkinson’s disease in mice via stimulation of glucagon like peptide-1. J. Neurol. Sci. 2017, 381, 176–181. [Google Scholar] [CrossRef]
- Zhang, Y.-G.; Wu, S.; Yi, J.; Xia, Y.; Jin, D.; Zhou, J.; Sun, J. Target Intestinal Microbiota to Alleviate Disease Progression in Amyotrophic Lateral Sclerosis. Clin. Ther. 2017, 39, 322–336. [Google Scholar] [CrossRef] [PubMed]
- Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Tsigalou, C.; Dalamaga, M. Probiotics, Prebiotics, Synbiotics, Postbiotics, and Obesity: Current Evidence, Controversies, and Perspectives. Curr. Obes. Rep. 2020, 9, 179–192. [Google Scholar] [CrossRef]
- Reid, G.; Younes, J.A.; Van der Mei, H.C.; Gloor, G.B.; Knight, R.; Busscher, H.J. Microbiota restoration: Natural and supplemented recovery of human microbial communities. Nat. Rev. Microbiol. 2011, 9, 27–38. [Google Scholar] [CrossRef]
- Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. Biomed. Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef]
- Abildgaard, A.; Elfving, B.; Hokland, M.; Wegener, G.; Lund, S. Probiotic treatment reduces depressive-like behaviour in rats independently of diet. Psychoneuroendocrinology 2017, 79, 40–48. [Google Scholar] [CrossRef]
- Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef]
- Suganya, K.; Koo, B.-S. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef] [PubMed]
- Rezaei Asl, Z.; Sepehri, G.; Salami, M. Probiotic treatment improves the impaired spatial cognitive performance and restores synaptic plasticity in an animal model of Alzheimer’s disease. Behav. Brain Res. 2019, 376, 112183. [Google Scholar] [CrossRef]
- Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; 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]
- Bonfili, L.; Cecarini, V.; Cuccioloni, M.; Angeletti, M.; Berardi, S.; Scarpona, S.; Rossi, G.; Eleuteri, A.M. SLAB51 Probiotic Formulation Activates SIRT1 Pathway Promoting Antioxidant and Neuroprotective Effects in an AD Mouse Model. Mol. Neurobiol. 2018, 55, 7987–8000. [Google Scholar] [CrossRef]
- Castelli, V.; d’Angelo, M.; Lombardi, F.; Alfonsetti, M.; Antonosante, A.; Catanesi, M.; Benedetti, E.; Palumbo, P.; Cifone, M.G.; Giordano, A.; et al. Effects of the probiotic formulation SLAB51 in in vitro and in vivo Parkinson’s disease models. Aging 2020, 12, 4641–4659. [Google Scholar] [CrossRef]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
- Tan, Q.; Orsso, C.E.; Deehan, E.C.; Kung, J.Y.; Tun, H.M.; Wine, E.; Madsen, K.L.; Zwaigenbaum, L.; Haqq, A.M. Probiotics, prebiotics, synbiotics, and fecal microbiota transplantation in the treatment of behavioral symptoms of autism spectrum disorder: A systematic review. Autism Res. 2021, 14, 1820–1836. [Google Scholar] [CrossRef]
- Pokusaeva, K.; Fitzgerald, G.F.; van Sinderen, D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. 2011, 6, 285–306. [Google Scholar] [CrossRef]
- Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement 2018, 4, 575–590. [Google Scholar] [CrossRef]
- Li, W.; Guo, J.; Shen, Y.; Huang, L.; Leng, B.; Fan, D.; Shui, L.; Chen, C. Probiotics, prebiotics, and synbiotics for the treatment of dementia: Protocol for a systematic review. Medicine 2020, 99, e18608. [Google Scholar] [CrossRef]
- Rocha, N.P.; de Miranda, A.S.; Teixeira, A.L. Insights into Neuroinflammation in Parkinson’s Disease: From Biomarkers to Anti-Inflammatory Based Therapies. Biomed. Res. Int. 2015, 2015, 628192. [Google Scholar] [CrossRef] [PubMed]
- Siniscalco, D.; Schultz, S.; Brigida, A.L.; Antonucci, N. Inflammation and Neuro-Immune Dysregulations in Autism Spectrum Disorders. Pharmaceuticals 2018, 11, 56. [Google Scholar] [CrossRef]
- Sun, J.; Liu, S.; Ling, Z.; Wang, F.; Ling, Y.; Gong, T.; Fang, N.; Ye, S.; Si, J.; Liu, J. Fructooligosaccharides Ameliorating Cognitive Deficits and Neurodegeneration in APP/PS1 Transgenic Mice through Modulating Gut Microbiota. J. Agric. Food Chem. 2019, 67, 3006–3017. [Google Scholar] [CrossRef]
- Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef]
- Artiss, J.D.; Brogan, K.; Brucal, M.; Moghaddam, M.; Jen, K.L.C. The effects of a new soluble dietary fiber on weight gain and selected blood parameters in rats. Metabolism 2006, 55, 195–202. [Google Scholar] [CrossRef]
- Yang, X.-D.; Wang, L.-K.; Wu, H.-Y.; Jiao, L. Effects of prebiotic galacto-oligosaccharide on postoperative cognitive dysfunction and neuroinflammation through targeting of the gut-brain axis. BMC Anesthesiol. 2018, 18, 177. [Google Scholar] [CrossRef]
- Marco, M.L.; Sanders, M.E.; Gänzle, M.; Arrieta, M.C.; Cotter, P.D.; De Vuyst, L.; Hill, C.; Holzapfel, W.; Lebeer, S.; Merenstein, D.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 196–208. [Google Scholar] [CrossRef]
- Chan, C.K.Y.; Tao, J.; Chan, O.S.; Li, H.-B.; Pang, H. Preventing Respiratory Tract Infections by Synbiotic Interventions: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2020, 11, 979–988. [Google Scholar] [CrossRef]
- Barichella, M.; Pacchetti, C.; Bolliri, C.; Cassani, E.; Iorio, L.; Pusani, C.; Pinelli, G.; Privitera, G.; Cesari, I.; Faierman, S.A.; et al. Probiotics and prebiotic fiber for constipation associated with Parkinson disease: An RCT. Neurology 2016, 87, 1274–1280. [Google Scholar] [CrossRef]
- Andreozzi, V.; Cuoco, S.; Balestrieri, M.; Fierro, F.; Ferrara, N.; Erro, R.; Di Filippo, M.; Barbella, G.; Memoli, M.C.; Silvestri, A.; et al. Synbiotic supplementation may globally improve non-motor symptoms in patients with stable Parkinson’s disease: Results from an open label single-arm study. Sci. Rep. 2024, 14, 23095. [Google Scholar] [CrossRef]
- Li, C.; Zhao, X.; Xu, H.; Liu, X.; He, Y.; Gu, J. NMN Synbiotics: A Multifaceted Therapeutic Approach for Alzheimer’s Disease. Neurochem. Res. 2024, 49, 2888–2896. [Google Scholar] [CrossRef] [PubMed]
- Cencic, A.; Chingwaru, W. The role of functional foods, nutraceuticals, and food supplements in intestinal health. Nutrients 2010, 2, 611–625. [Google Scholar] [CrossRef] [PubMed]
- Westfall, S.; Lomis, N.; Prakash, S. A novel synbiotic delays Alzheimer’s disease onset via combinatorial gut-brain-axis signaling in Drosophila melanogaster. PLoS ONE 2019, 14, e0214985. [Google Scholar] [CrossRef]
- Mohanty, M.; Mohanty, P.S. Molecular docking in organic, inorganic, and hybrid systems: A tutorial review. Monatsh Chem. 2023, 6, 683–707. [Google Scholar] [CrossRef] [PubMed]
- Vinderola, G.; Sanders, M.E.; Salminen, S. The Concept of Postbiotics. Foods 2022, 11, 1077. [Google Scholar] [CrossRef]
- Scott, E.; De Paepe, K.; Van de Wiele, T. Postbiotics and Their Health Modulatory Biomolecules. Biomolecules 2022, 12, 1640. [Google Scholar] [CrossRef]
- Prajapati, N.; Patel, J.; Singh, S.; Yadav, V.K.; Joshi, C.; Patani, A.; Prajapati, D.; Sahoo, D.K.; Patel, A. Postbiotic production: Harnessing the power of microbial metabolites for health applications. Front. Microbiol. 2023, 14, 1306192. [Google Scholar] [CrossRef]
- Zhu, M.; Liu, X.; Ye, Y.; Yan, X.; Cheng, Y.; Zhao, L.; Chen, F.; Ling, Z. Gut Microbiota: A Novel Therapeutic Target for Parkinson’s Disease. Front. Immunol. 2022, 13, 937555. [Google Scholar] [CrossRef]
- Bashir, B.; Alam, S.; Khandale, N.; Birla, D.; Vishwas, S.; Pandey, N.K.; Gupta, G.; Paudel, K.R.; Dureja, H.; Kumar, P.; et al. Opening avenues for treatment of neurodegenerative disease using post-biotics: Breakthroughs and bottlenecks in clinical translation. Ageing Res. Rev. 2024, 95, 102236. [Google Scholar] [CrossRef]
- Rosenfeld, C.S. Microbiome Disturbances and Autism Spectrum Disorders. Drug Metab. Dispos. 2015, 43, 1557–1571. [Google Scholar] [CrossRef]
- Braga, J.D.; Thongngam, M.; Kumrungsee, T. Gamma-aminobutyric acid as a potential postbiotic mediator in the gut-brain axis. NPJ Sci. Food 2024, 8, 16. [Google Scholar] [CrossRef] [PubMed]
- Mincic, A.M.; Antal, M.; Filip, L.; Miere, D. Modulation of gut microbiome in the treatment of neurodegenerative diseases: A systematic review. Clin. Nutr. 2024, 43, 1832–1849. [Google Scholar] [CrossRef]
- Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef]
- Pellegrini, C.; Fornai, M.; D’Antongiovanni, V.; Antonioli, L.; Bernardini, N.; Derkinderen, P. The intestinal barrier in disorders of the central nervous system. Lancet Gastroenterol. Hepatol. 2023, 8, 66–80. [Google Scholar] [CrossRef]
- Fasano, A. All disease begins in the (leaky) gut: Role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Res 2020, 9, F1000 Faculty Rev-69. [Google Scholar] [CrossRef]
- Theoharides, T.C.; Asadi, S.; Patel, A.B. Focal brain inflammation and autism. J. Neuroinflammation 2013, 10, 46. [Google Scholar] [CrossRef]
- Zeisel, M.B.; Dhawan, P.; Baumert, T.F. Tight junction proteins in gastrointestinal and liver disease. Gut 2019, 68, 547–561. [Google Scholar] [CrossRef]
- La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the Gut Immune System: Indirect Regulation. Probiotics Antimicrob. Proteins 2018, 10, 11–21. [Google Scholar] [CrossRef]
- Gilbert, J.A.; Krajmalnik-Brown, R.; Porazinska, D.L.; Weiss, S.J.; Knight, R. Toward effective probiotics for autism and other neurodevelopmental disorders. Cell 2013, 155, 1446–1448. [Google Scholar] [CrossRef]
- Wang, D.D.; Nguyen, L.H.; Li, Y.; Yan, Y.; Ma, W.; Rinott, E.; Ivey, K.L.; Shai, I.; Willett, W.C.; Hu, F.B.; et al. The gut microbiome modulates the protective association between a Mediterranean diet and cardiometabolic disease risk. Nat. Med. 2021, 27, 333–343. [Google Scholar] [CrossRef]
- Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Li, N.; Yang, J.-J.; Zhao, D.-M.; Chen, B.; Zhang, G.-Q.; Chen, S.; Cao, R.-F.; Yu, H.; Zhao, C.-Y.; et al. Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacol. Res. 2020, 157, 104784. [Google Scholar] [CrossRef] [PubMed]
- Hall, D.A.; Voigt, R.M.; Cantu-Jungles, T.M.; Hamaker, B.; Engen, P.A.; Shaikh, M.; Raeisi, S.; Green, S.J.; Naqib, A.; Forsyth, C.B.; et al. An open label, non-randomized study assessing a prebiotic fiber intervention in a small cohort of Parkinson’s disease participants. Nat. Commun. 2023, 14, 926. [Google Scholar] [CrossRef]
- Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef]
- Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef]
- Fock, E.; Parnova, R. Mechanisms of Blood-Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells 2023, 12, 657. [Google Scholar] [CrossRef]
- Yan, F.; Polk, D.B. Probiotics and immune health. Curr. Opin. Gastroenterol. 2011, 27, 496–501. [Google Scholar] [CrossRef]
- Hardy, H.; Harris, J.; Lyon, E.; Beal, J.; Foey, A.D. Probiotics, prebiotics and immunomodulation of gut mucosal defences: Homeostasis and immunopathology. Nutrients 2013, 5, 1869–1912. [Google Scholar] [CrossRef]
- Shokryazdan, P.; Faseleh Jahromi, M.; Navidshad, B.; Liang, J.B. Effects of prebiotics on immune system and cytokine expression. Med. Microbiol. Immunol. 2017, 206, 1–9. [Google Scholar] [CrossRef]
- Camara-Lemarroy, C.R.; Metz, L.; Meddings, J.B.; Sharkey, K.A.; Wee Yong, V. The intestinal barrier in multiple sclerosis: Implications for pathophysiology and therapeutics. Brain 2018, 141, 1900–1916. [Google Scholar] [CrossRef]
- Hoyles, L.; Pontifex, M.G.; Rodriguez-Ramiro, I.; Anis-Alavi, M.A.; Jelane, K.S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A.L.; Carding, S.R.; et al. Regulation of blood-brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235. [Google Scholar] [CrossRef] [PubMed]
- Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome-host systems interactions: Protective effects of propionate upon the blood-brain barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef] [PubMed]
- Dolkar, P.; Deyang, T.; Anand, N.; Rathipriya, A.G.; Hediyal, T.A.; Chandrasekaran, V.; Krishnamoorthy, N.K.; Gorantla, V.R.; Bishir, M.; Rashan, L.; et al. Trimethylamine-N-oxide and cerebral stroke risk: A review. Neurobiol. Dis. 2024, 192, 106423. [Google Scholar] [CrossRef]
- Rivera, C.A.; Lennon-Duménil, A.-M. Gut immune cells and intestinal niche imprinting. Semin. Cell Dev. Biol. 2023, 150–151, 50–57. [Google Scholar] [CrossRef]
- Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef]
- Chitnis, T.; Weiner, H.L. CNS inflammation and neurodegeneration. J. Clin. Investig. 2017, 127, 3577–3587. [Google Scholar] [CrossRef]
- Gjelstrup, M.C.; Stilund, M.; Petersen, T.; Møller, H.J.; Petersen, E.L.; Christensen, T. Subsets of activated monocytes and markers of inflammation in incipient and progressed multiple sclerosis. Immunol. Cell Biol. 2018, 96, 160–174. [Google Scholar] [CrossRef]
- Yin, J.; Valin, K.L.; Dixon, M.L.; Leavenworth, J.W. The Role of Microglia and Macrophages in CNS Homeostasis, Autoimmunity, and Cancer. J. Immunol. Res. 2017, 2017, 5150678. [Google Scholar] [CrossRef]
- Schachtsiek, M.; Hammes, W.P.; Hertel, C. Characterization of Lactobacillus coryniformis DSM 20001T surface protein Cpf mediating coaggregation with and aggregation among pathogens. Appl. Environ. Microbiol. 2004, 70, 7078–7085. [Google Scholar] [CrossRef]
- Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef]
- Rothhammer, V.; Quintana, F.J. The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 2019, 19, 184–197. [Google Scholar] [CrossRef] [PubMed]
- Fukumoto, S.; Toshimitsu, T.; Matsuoka, S.; Maruyama, A.; Oh-Oka, K.; Takamura, T.; Nakamura, Y.; Ishimaru, K.; Fujii-Kuriyama, Y.; Ikegami, S.; et al. Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis. Immunol. Cell Biol. 2014, 92, 460–465. [Google Scholar] [CrossRef] [PubMed]
- Marafini, I.; Monteleone, I.; Laudisi, F.; Monteleone, G. Aryl Hydrocarbon Receptor Signalling in the Control of Gut Inflammation. Int. J. Mol. Sci. 2024, 25, 4527. [Google Scholar] [CrossRef] [PubMed]
- Vulevic, J.; Drakoularakou, A.; Yaqoob, P.; Tzortzis, G.; Gibson, G.R. Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers. Am. J. Clin. Nutr. 2008, 88, 1438–1446. [Google Scholar] [CrossRef]
- Dargahi, N.; Matsoukas, J.; Apostolopoulos, V. Streptococcus thermophilus ST285 Alters Pro-Inflammatory to Anti-Inflammatory Cytokine Secretion against Multiple Sclerosis Peptide in Mice. Brain Sci. 2020, 10, 126. [Google Scholar] [CrossRef]
- Kim, C.H. Control of lymphocyte functions by gut microbiota-derived short-chain fatty acids. Cell Mol. Immunol. 2021, 18, 1161–1171. [Google Scholar] [CrossRef]
- Kim, C.H. Complex regulatory effects of gut microbial short-chain fatty acids on immune tolerance and autoimmunity. Cell Mol. Immunol. 2023, 20, 341–350. [Google Scholar] [CrossRef]
- Rosser, E.C.; Piper, C.J.M.; Matei, D.E.; Blair, P.A.; Rendeiro, A.F.; Orford, M.; Alber, D.G.; Krausgruber, T.; Catalan, D.; Klein, N.; et al. Microbiota-Derived Metabolites Suppress Arthritis by Amplifying Aryl-Hydrocarbon Receptor Activation in Regulatory B Cells. Cell Metab. 2020, 31, 837–851.e10. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Mucida, D.; Park, Y.; Kim, G.; Turovskaya, O.; Scott, I.; Kronenberg, M.; Cheroutre, H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007, 317, 256–260. [Google Scholar] [CrossRef] [PubMed]
- Haghikia, A.; Jörg, S.; Duscha, A.; Berg, J.; Manzel, A.; Waschbisch, A.; Hammer, A.; Lee, D.-H.; May, C.; Wilck, N.; et al. Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 2016, 44, 951–953. [Google Scholar] [CrossRef]
- Nimgampalle, M.; Chakravarthy, H.; Sharma, S.; Shree, S.; Bhat, A.R.; Pradeepkiran, J.A.; Devanathan, V. Neurotransmitter systems in the etiology of major neurological disorders: Emerging insights and therapeutic implications. Ageing Res. Rev. 2023, 89, 101994. [Google Scholar] [CrossRef]
- Forsythe, P.; Bienenstock, J.; Kunze, W.A. Vagal pathways for microbiome-brain-gut axis communication. Adv. Exp. Med. Biol. 2014, 817, 115–133. [Google Scholar]
- Dong, X.-L.; Wang, X.; Liu, F.; Liu, X.; Du, Z.-R.; Li, R.W.; Xue, C.-H.; Wong, K.-H.; Wong, W.-T.; Zhao, Q.; et al. Polymannuronic acid prevents dopaminergic neuronal loss via brain-gut-microbiota axis in Parkinson’s disease model. Int. J. Biol. Macromol. 2020, 164, 994–1005. [Google Scholar] [CrossRef]
- Conn, K.A.; Borsom, E.M.; Cope, E.K. Implications of microbe-derived ɣ-aminobutyric acid (GABA) in gut and brain barrier integrity and GABAergic signaling in Alzheimer’s disease. Gut Microbes 2024, 16, 2371950. [Google Scholar] [CrossRef]
- Sun, H.; Zhao, F.; Liu, Y.; Ma, T.; Jin, H.; Quan, K.; Leng, B.; Zhao, J.; Yuan, X.; Li, Z.; et al. Probiotics synergized with conventional regimen in managing Parkinson’s disease. NPJ Parkin. Dis. 2022, 8, 62. [Google Scholar] [CrossRef]
- Borovikova, L.V.; Ivanova, S.; Zhang, M.; Yang, H.; Botchkina, G.I.; Watkins, L.R.; Wang, H.; Abumrad, N.; Eaton, J.W.; Tracey, K.J. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000, 405, 458–462. [Google Scholar] [CrossRef]
- Reale, M.; Costantini, E. Cholinergic Modulation of the Immune System in Neuroinflammatory Diseases. Diseases 2021, 9, 29. [Google Scholar] [CrossRef]
- Lane, R.M.; Potkin, S.G.; Enz, A. Targeting acetylcholinesterase and butyrylcholinesterase in dementia. Int. J. Neuropsychopharmacol. 2006, 9, 101–124. [Google Scholar] [CrossRef]
- Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, KC01–KC05. [Google Scholar] [CrossRef] [PubMed]
- Sun, J.; Li, H.; Jin, Y.; Yu, J.; Mao, S.; Su, K.-P.; Ling, Z.; Liu, J. Probiotic Clostridium butyricum ameliorated motor deficits in a mouse model of Parkinson’s disease via gut microbiota-GLP-1 pathway. Brain Behav. Immun. 2021, 91, 703–715. [Google Scholar] [CrossRef] [PubMed]
- Weninger, S.N.; Herman, C.; Meyer, R.K.; Beauchemin, E.T.; Kangath, A.; Lane, A.I.; Martinez, T.M.; Hasneen, T.; Jaramillo, S.A.; Lindsey, J.; et al. Oligofructose improves small intestinal lipid-sensing mechanisms via alterations to the small intestinal microbiota. Microbiome 2023, 11, 169. [Google Scholar] [CrossRef] [PubMed]
- De Angelis, M.; Francavilla, R.; Piccolo, M.; De Giacomo, A.; Gobbetti, M. Autism spectrum disorders and intestinal microbiota. Gut Microbes 2015, 6, 207–213. [Google Scholar] [CrossRef]
- Winek, K.; Dirnagl, U.; Meisel, A. The Gut Microbiome as Therapeutic Target in Central Nervous System Diseases: Implications for Stroke. Neurotherapeutics 2016, 13, 762–774. [Google Scholar] [CrossRef]
- 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]
- Sandler, R.H.; Finegold, S.M.; Bolte, E.R.; Buchanan, C.P.; Maxwell, A.P.; Väisänen, M.L.; Nelson, M.N.; Wexler, H.M. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J. Child. Neurol. 2000, 15, 429–435. [Google Scholar] [CrossRef]
- Adams, J.B.; Johansen, L.J.; Powell, L.D.; Quig, D.; Rubin, R.A. Gastrointestinal flora and gastrointestinal status in children with autism—Comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011, 11, 22. [Google Scholar] [CrossRef]
- Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl. Environ. Microbiol. 2011, 77, 6718–6721. [Google Scholar] [CrossRef]
- Berding, K.; Donovan, S.M. Microbiome and nutrition in autism spectrum disorder: Current knowledge and research needs. Nutr. Rev. 2016, 74, 723–736. [Google Scholar] [CrossRef]
- Markowiak, P.; Śliżewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef] [PubMed]
- Shadnoush, M.; Hosseini, R.S.; Khalilnezhad, A.; Navai, L.; Goudarzi, H.; Vaezjalali, M. Effects of Probiotics on Gut Microbiota in Patients with Inflammatory Bowel Disease: A Double-blind, Placebo-controlled Clinical Trial. Korean J. Gastroenterol. 2015, 65, 215–221. [Google Scholar] [CrossRef]
- Erhardt, R.; Harnett, J.E.; Steels, E.; Steadman, K.J. Functional constipation and the effect of prebiotics on the gut microbiota: A review. Br. J. Nutr. 2023, 130, 1015–1023. [Google Scholar] [CrossRef]
- Shang, Q.; Shan, X.; Cai, C.; Hao, J.; Li, G.; Yu, G. Dietary fucoidan modulates the gut microbiota in mice by increasing the abundance of Lactobacillus and Ruminococcaceae. Food Funct. 2016, 7, 3224–3232. [Google Scholar] [CrossRef]
- Wang, H.; Zhang, X.; Wang, S.; Li, H.; Lu, Z.; Shi, J.; Xu, Z. Mannan-oligosaccharide modulates the obesity and gut microbiota in high-fat diet-fed mice. Food Funct. 2018, 9, 3916–3929. [Google Scholar] [CrossRef]
- Rout, M.; Kar, D.M.; Dubey, D.; Kispotta, S.; Sarangi, P.; Prusty, S.K. Neuroprotective effect of Bacillus subtilis in haloperidol induced rat model, targeting the microbiota-gut-brain axis. J. Mol. Histol. 2024, 56, 18. [Google Scholar] [CrossRef]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef]
- Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef]
- Welly, R.J.; Liu, T.-W.; Zidon, T.M.; Rowles, J.L.; Park, Y.-M.; Smith, T.N.; Swanson, K.S.; Padilla, J.; Vieira-Potter, V.J. Comparison of Diet versus Exercise on Metabolic Function and Gut Microbiota in Obese Rats. Med. Sci. Sports Exerc. 2016, 48, 1688–1698. [Google Scholar] [CrossRef]
- Monda, V.; Villano, I.; Messina, A.; Valenzano, A.; Esposito, T.; Moscatelli, F.; Viggiano, A.; Cibelli, G.; Chieffi, S.; Monda, M.; et al. Exercise Modifies the Gut Microbiota with Positive Health Effects. Oxid. Med. Cell Longev. 2017, 2017, 3831972. [Google Scholar] [CrossRef]
- Allen, J.M.; Mailing, L.J.; Cohrs, J.; Salmonson, C.; Fryer, J.D.; Nehra, V.; Hale, V.L.; Kashyap, P.; White, B.A.; Woods, J.A. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes 2018, 9, 115–130. [Google Scholar] [CrossRef] [PubMed]
- Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef]
- Batacan, R.B.; Fenning, A.S.; Dalbo, V.J.; Scanlan, A.T.; Duncan, M.J.; Moore, R.J.; Stanley, D. A gut reaction: The combined influence of exercise and diet on gastrointestinal microbiota in rats. J. Appl. Microbiol. 2017, 122, 1627–1638. [Google Scholar] [CrossRef]
- Campbell, S.C.; Wisniewski, P.J.; Noji, M.; McGuinness, L.R.; Häggblom, M.M.; Lightfoot, S.A.; Joseph, L.B.; Kerkhof, L.J. The Effect of Diet and Exercise on Intestinal Integrity and Microbial Diversity in Mice. PLoS ONE 2016, 11, e0150502. [Google Scholar] [CrossRef]
- Matsumoto, M.; Inoue, R.; Tsukahara, T.; Ushida, K.; Chiji, H.; Matsubara, N.; Hara, H. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Biosci. Biotechnol. Biochem. 2008, 72, 572–576. [Google Scholar] [CrossRef]
- Queipo-Ortuño, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.; Casanueva, F.; Tinahones, F.J. Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 2013, 8, e65465. [Google Scholar] [CrossRef]
- Cerdá, B.; Pérez, M.; Pérez-Santiago, J.D.; Tornero-Aguilera, J.F.; González-Soltero, R.; Larrosa, M. Gut Microbiota Modification: Another Piece in the Puzzle of the Benefits of Physical Exercise in Health? Front. Physiol. 2016, 7, 51. [Google Scholar] [CrossRef]
- Lavie, C.J.; Church, T.S.; Milani, R.V.; Earnest, C.P. Impact of physical activity, cardiorespiratory fitness, and exercise training on markers of inflammation. J. Cardiopulm. Rehabil. Prev. 2011, 31, 137–145. [Google Scholar] [CrossRef]
- Graham, L.C.; Grabowska, W.A.; Chun, Y.; Risacher, S.L.; Philip, V.M.; Saykin, A.J.; Sukoff Rizzo, S.J.; Howell, G.R. Exercise prevents obesity-induced cognitive decline and white matter damage in mice. Neurobiol. Aging 2019, 80, 154–172. [Google Scholar] [CrossRef]
- McGurran, H.; Glenn, J.M.; Madero, E.N.; Bott, N.T. Prevention and Treatment of Alzheimer’s Disease: Biological Mechanisms of Exercise. J. Alzheimers Dis. 2019, 69, 311–338. [Google Scholar] [CrossRef]
- Rasmussen, P.; Brassard, P.; Adser, H.; Pedersen, M.V.; Leick, L.; Hart, E.; Secher, N.H.; Pedersen, B.K.; Pilegaard, H. Evidence for a release of brain-derived neurotrophic factor from the brain during exercise. Exp. Physiol. 2009, 94, 1062–1069. [Google Scholar] [CrossRef] [PubMed]
- Molteni, R.; Wu, A.; Vaynman, S.; Ying, Z.; Barnard, R.J.; Gómez-Pinilla, F. Exercise reverses the harmful effects of consumption of a high-fat diet on synaptic and behavioral plasticity associated to the action of brain-derived neurotrophic factor. Neuroscience 2004, 123, 429–440. [Google Scholar] [CrossRef] [PubMed]
- Yu, F.; Vock, D.M.; Zhang, L.; Salisbury, D.; Nelson, N.W.; Chow, L.S.; Smith, G.; Barclay, T.R.; Dysken, M.; Wyman, J.F. Cognitive Effects of Aerobic Exercise in Alzheimer’s Disease: A Pilot Randomized Controlled Trial. J. Alzheimers Dis. 2021, 80, 233–244. [Google Scholar] [CrossRef]
- Meng, L.; Li, X.; Li, C.; Tsang, R.C.C.; Chen, Y.; Ge, Y.; Gao, Q. Effects of Exercise in Patients With Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Am. J. Phys. Med. Rehabil. 2020, 99, 801–810. [Google Scholar] [CrossRef]
- Jansen, A.E.; Koop, M.M.; Rosenfeldt, A.B.; Alberts, J.L. High intensity aerobic exercise improves bimanual coordination of grasping forces in Parkinson’s disease. Parkin. Relat. Disord. 2021, 87, 13–19. [Google Scholar] [CrossRef]
- Dinu, M.; Pagliai, G.; Casini, A.; Sofi, F. Mediterranean diet and multiple health outcomes: An umbrella review of meta-analyses of observational studies and randomised trials. Eur. J. Clin. Nutr. 2018, 72, 30–43. [Google Scholar] [CrossRef]
- Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef]
- Abenavoli, L.; Gambardella, M.L.; Scarlata, G.G.M.; Lenci, I.; Baiocchi, L.; Luzza, F. The Many Faces of Metabolic Dysfunction-Associated Fatty Liver Disease Treatment: From the Mediterranean Diet to Fecal Microbiota Transplantation. Medicina 2024, 60, 563. [Google Scholar] [CrossRef]
- Bernardes, D.; Brambilla, R.; Bracchi-Ricard, V.; Karmally, S.; Dellarole, A.; Carvalho-Tavares, J.; Bethea, J.R. Prior regular exercise improves clinical outcome and reduces demyelination and axonal injury in experimental autoimmune encephalomyelitis. J. Neurochem. 2016, 136 (Suppl. 1), 63–73. [Google Scholar] [CrossRef]
- Xie, Y.; Li, Z.; Wang, Y.; Xue, X.; Ma, W.; Zhang, Y.; Wang, J. Effects of moderate- versus high- intensity swimming training on inflammatory and CD4+ T cell subset profiles in experimental autoimmune encephalomyelitis mice. J. Neuroimmunol. 2019, 328, 60–67. [Google Scholar] [CrossRef]
- Mastorakos, G.; Pavlatou, M.; Diamanti-Kandarakis, E.; Chrousos, G.P. Exercise and the stress system. Hormones 2005, 4, 73–89. [Google Scholar]
- Velloso, L.A.; Donato, J. Growth Hormone, Hypothalamic Inflammation, and Aging. J. Obes. Metab. Syndr. 2024, 33, 302–313. [Google Scholar] [CrossRef]
- Costanza, M.; Pedotti, R. Prolactin: Friend or Foe in Central Nervous System Autoimmune Inflammation? Int. J. Mol. Sci. 2016, 17, 2026. [Google Scholar] [CrossRef] [PubMed]
- Kurgan, N.; Noaman, N.; Pergande, M.R.; Cologna, S.M.; Coorssen, J.R.; Klentrou, P. Changes to the Human Serum Proteome in Response to High Intensity Interval Exercise: A Sequential Top-Down Proteomic Analysis. Front. Physiol. 2019, 10, 362. [Google Scholar] [CrossRef]
- da Luz Scheffer, D.; Latini, A. Exercise-induced immune system response: Anti-inflammatory status on peripheral and central organs. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165823. [Google Scholar]
- Campbell, J.P.; Turner, J.E. Debunking the Myth of Exercise-Induced Immune Suppression: Redefining the Impact of Exercise on Immunological Health Across the Lifespan. Front. Immunol. 2018, 9, 648. [Google Scholar] [CrossRef]
- Gentile, A.; Musella, A.; De Vito, F.; Rizzo, F.R.; Fresegna, D.; Bullitta, S.; Vanni, V.; Guadalupi, L.; Stampanoni Bassi, M.; Buttari, F.; et al. Immunomodulatory Effects of Exercise in Experimental Multiple Sclerosis. Front. Immunol. 2019, 10, 2197. [Google Scholar] [CrossRef]
- Shandilya, S.; Kumar, S.; Kumar Jha, N.; Kumar Kesari, K.; Ruokolainen, J. Interplay of gut microbiota and oxidative stress: Perspective on neurodegeneration and neuroprotection. J. Adv. Res. 2022, 38, 223–244. [Google Scholar] [CrossRef]
- Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar]
- Hood, D.A.; Memme, J.M.; Oliveira, A.N.; Triolo, M. Maintenance of Skeletal Muscle Mitochondria in Health, Exercise, and Aging. Annu. Rev. Physiol. 2019, 81, 19–41. [Google Scholar] [CrossRef]
- Memme, J.M.; Erlich, A.T.; Phukan, G.; Hood, D.A. Exercise and mitochondrial health. J. Physiol. 2021, 599, 803–817. [Google Scholar] [CrossRef] [PubMed]
- Souza, P.S.; Gonçalves, E.D.; Pedroso, G.S.; Farias, H.R.; Junqueira, S.C.; Marcon, R.; Tuon, T.; Cola, M.; Silveira, P.C.L.; Santos, A.R.; et al. Physical Exercise Attenuates Experimental Autoimmune Encephalomyelitis by Inhibiting Peripheral Immune Response and Blood-Brain Barrier Disruption. Mol. Neurobiol. 2017, 54, 4723–4737. [Google Scholar] [CrossRef]
- Lin, B.; Wu, T.; Nasb, M.; Li, Z.; Chen, N. Regular exercise alleviates metabolic dysfunction-associated steatohepatitis through rescuing mitochondrial oxidative stress and dysfunction in liver. Free Radic. Biol. Med. 2025, 230, 163–176. [Google Scholar] [CrossRef]
- Bloomer, R.J.; Schilling, B.K.; Karlage, R.E.; Ledoux, M.S.; Pfeiffer, R.F.; Callegari, J. Effect of resistance training on blood oxidative stress in Parkinson disease. Med. Sci. Sports Exerc. 2008, 40, 1385–1389. [Google Scholar] [CrossRef]
- Tuon, T.; Valvassori, S.S.; Lopes-Borges, J.; Luciano, T.; Trom, C.B.; Silva, L.A.; Quevedo, J.; Souza, C.T.; Lira, F.S.; Pinho, R.A. Physical training exerts neuroprotective effects in the regulation of neurochemical factors in an animal model of Parkinson’s disease. Neuroscience 2012, 227, 305–312. [Google Scholar] [CrossRef]
- Lin, T.-W.; Kuo, Y.-M. Exercise benefits brain function: The monoamine connection. Brain Sci. 2013, 3, 39–53. [Google Scholar] [CrossRef]
- Meeusen, R.; De Meirleir, K. Exercise and brain neurotransmission. Sports Med. 1995, 20, 160–188. [Google Scholar] [CrossRef]
- Mills, K.C. Serotonin syndrome. A clinical update. Crit. Care Clin. 1997, 13, 763–783. [Google Scholar] [CrossRef]
- Jian, H.; Li, R.; Huang, X.; Li, J.; Li, Y.; Ma, J.; Zhu, M.; Dong, X.; Yang, H.; Zou, X. Branched-chain amino acids alleviate NAFLD via inhibiting de novo lipogenesis and activating fatty acid β-oxidation in laying hens. Redox Biol. 2024, 77, 103385. [Google Scholar] [CrossRef]
- Patrick, R.P.; Ames, B.N. Vitamin D and the omega-3 fatty acids control serotonin synthesis and action, part 2: Relevance for ADHD, bipolar disorder, schizophrenia, and impulsive behavior. FASEB J. 2015, 29, 2207–2222. [Google Scholar] [CrossRef] [PubMed]
- Chennaoui, M.; Grimaldi, B.; Fillion, M.P.; Bonnin, A.; Drogou, C.; Fillion, G.; Guezennec, C.Y. Effects of physical training on functional activity of 5-HT1B receptors in rat central nervous system: Role of 5-HT-moduline. Naunyn Schmiedebergs Arch. Pharmacol. 2000, 361, 600–604. [Google Scholar] [CrossRef]
- Franco, R.; Reyes-Resina, I.; Navarro, G. Dopamine in Health and Disease: Much More Than a Neurotransmitter. Biomedicines 2021, 9, 109. [Google Scholar] [CrossRef] [PubMed]
- Rasheed, N.; Alghasham, A. Central dopaminergic system and its implications in stress-mediated neurological disorders and gastric ulcers: Short review. Adv. Pharmacol. Sci. 2012, 2012, 182671. [Google Scholar] [CrossRef]
- Sutoo, D.E.; Akiyama, K. Regulation of brain function by exercise. Neurobiol. Dis. 2003, 13, 1–14. [Google Scholar] [CrossRef]
- Das, G.; Gopalakrishnan, A.; Faisal, M.; Mallick, B.N. Stimulatory role of calcium in rapid eye movement sleep deprivation-induced noradrenaline-mediated increase in Na-K-ATPase activity in rat brain. Neuroscience 2008, 155, 76–89. [Google Scholar] [CrossRef] [PubMed]
- de Los Reyes, T.; Casas-Tintó, S. Neural functions of small heat shock proteins. Neural Regen. Res. 2022, 17, 512–515. [Google Scholar]
- Dello Russo, C.; Boullerne, A.I.; Gavrilyuk, V.; Feinstein, D.L. Inhibition of microglial inflammatory responses by norepinephrine: Effects on nitric oxide and interleukin-1beta production. J. Neuroinflamm. 2004, 1, 9. [Google Scholar] [CrossRef]
- Feinstein, D.L.; Heneka, M.T.; Gavrilyuk, V.; Dello Russo, C.; Weinberg, G.; Galea, E. Noradrenergic regulation of inflammatory gene expression in brain. Neurochem. Int. 2002, 41, 357–365. [Google Scholar] [CrossRef]
- Klotz, L.; Sastre, M.; Kreutz, A.; Gavrilyuk, V.; Klockgether, T.; Feinstein, D.L.; Heneka, M.T. Noradrenaline induces expression of peroxisome proliferator activated receptor gamma (PPARgamma) in murine primary astrocytes and neurons. J. Neurochem. 2003, 86, 907–916. [Google Scholar] [CrossRef]
- Ghasemi, M.; Mehranfard, N. Neuroprotective actions of norepinephrine in neurological diseases. Pflugers Arch. 2024, 476, 1703–1725. [Google Scholar] [CrossRef]
- Ross, R.E.; VanDerwerker, C.J.; Saladin, M.E.; Gregory, C.M. The role of exercise in the treatment of depression: Biological underpinnings and clinical outcomes. Mol. Psychiatry 2023, 28, 298–328. [Google Scholar] [CrossRef] [PubMed]
- Birling, M.C.; Price, J. Influence of growth factors on neuronal differentiation. Curr. Opin. Cell Biol. 1995, 7, 878–884. [Google Scholar] [CrossRef]
- Thoenen, H. Neurotrophins and neuronal plasticity. Science 1995, 270, 593–598. [Google Scholar] [CrossRef]
- Lewin, G.R.; Barde, Y.A. Physiology of the neurotrophins. Annu. Rev. Neurosci. 1996, 19, 289–317. [Google Scholar] [CrossRef]
- Huang, E.J.; Reichardt, L.F. Neurotrophins: Roles in neuronal development and function. Annu. Rev. Neurosci. 2001, 24, 677–736. [Google Scholar] [CrossRef]
- Dinoff, A.; Herrmann, N.; Swardfager, W.; Liu, C.S.; Sherman, C.; Chan, S.; Lanctôt, K.L. The Effect of Exercise Training on Resting Concentrations of Peripheral Brain-Derived Neurotrophic Factor (BDNF): A Meta-Analysis. PLoS ONE 2016, 11, e0163037. [Google Scholar] [CrossRef]
- Peng, S.; Wuu, J.; Mufson, E.J.; Fahnestock, M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J. Neurochem. 2005, 93, 1412–1421. [Google Scholar] [CrossRef]
- Qin, X.Y.; Cao, C.; Cawley, N.X.; Liu, T.T.; Yuan, J.; Loh, Y.P.; Cheng, Y. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: A meta-analysis study (N = 7277). Mol. Psychiatry 2017, 22, 312–320. [Google Scholar] [CrossRef]
- Mattson, M.P. Evolutionary aspects of human exercise—Born to run purposefully. Ageing Res. Rev. 2012, 11, 347–352. [Google Scholar] [CrossRef]
- De la Rosa, A.; Olaso-Gonzalez, G.; Arc-Chagnaud, C.; Millan, F.; Salvador-Pascual, A.; García-Lucerga, C.; Blasco-Lafarga, C.; Garcia-Dominguez, E.; Carretero, A.; Correas, A.G.; et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J. Sport. Health Sci. 2020, 9, 394–404. [Google Scholar] [CrossRef]
- Fiatarone Singh, M.A.; Gates, N.; Saigal, N.; Wilson, G.C.; Meiklejohn, J.; Brodaty, H.; Wen, W.; Singh, N.; Baune, B.T.; Suo, C.; et al. The Study of Mental and Resistance Training (SMART) study—Resistance training and/or cognitive training in mild cognitive impairment: A randomized, double-blind, double-sham controlled trial. J. Am. Med. Dir. Assoc. 2014, 15, 873–880. [Google Scholar] [CrossRef] [PubMed]
- Lin, T.-W.; Shih, Y.-H.; Chen, S.-J.; Lien, C.-H.; Chang, C.-Y.; Huang, T.-Y.; Chen, S.-H.; Jen, C.J.; Kuo, Y.-M. Running exercise delays neurodegeneration in amygdala and hippocampus of Alzheimer’s disease (APP/PS1) transgenic mice. Neurobiol. Learn. Mem. 2015, 118, 189–197. [Google Scholar] [CrossRef] [PubMed]
- Padmakumar, S.; Taha, M.S.; Kadakia, E.; Bleier, B.S.; Amiji, M.M. Delivery of neurotrophic factors in the treatment of age-related chronic neurodegenerative diseases. Expert. Opin. Drug Deliv. 2020, 17, 323–340. [Google Scholar] [CrossRef]
- Klein, R.L.; Lewis, M.H.; Muzyczka, N.; Meyer, E.M. Prevention of 6-hydroxydopamine-induced rotational behavior by BDNF somatic gene transfer. Brain Res. 1999, 847, 314–320. [Google Scholar] [CrossRef]
- Sun, M.; Kong, L.; Wang, X.; Lu, X.-g.; Gao, Q.; Geller, A.I. Comparison of the capability of GDNF, BDNF, or both, to protect nigrostriatal neurons in a rat model of Parkinson’s disease. Brain Res. 2005, 1052, 119–129. [Google Scholar] [CrossRef]
- Zhu, G.; Li, J.; He, L.; Wang, X.; Hong, X. MPTP-induced changes in hippocampal synaptic plasticity and memory are prevented by memantine through the BDNF-TrkB pathway. Br. J. Pharmacol. 2015, 172, 2354–2368. [Google Scholar] [CrossRef]
- Ashcroft, S.K.; Ironside, D.D.; Johnson, L.; Kuys, S.S.; Thompson-Butel, A.G. Effect of Exercise on Brain-Derived Neurotrophic Factor in Stroke Survivors: A Systematic Review and Meta-Analysis. Stroke 2022, 53, 3706–3716. [Google Scholar] [CrossRef]
- Vaynman, S.; Ying, Z.; Gomez-Pinilla, F. Interplay between brain-derived neurotrophic factor and signal transduction modulators in the regulation of the effects of exercise on synaptic-plasticity. Neuroscience 2003, 122, 647–657. [Google Scholar] [CrossRef]
- Cobianchi, S.; Arbat-Plana, A.; Lopez-Alvarez, V.M.; Navarro, X. Neuroprotective Effects of Exercise Treatments After Injury: The Dual Role of Neurotrophic Factors. Curr. Neuropharmacol. 2017, 15, 495–518. [Google Scholar] [CrossRef]
- Sampaio, T.B.; Savall, A.S.; Gutierrez, M.E.Z.; Pinton, S. Neurotrophic factors in Alzheimer’s and Parkinson’s diseases: Implications for pathogenesis and therapy. Neural Regen. Res. 2017, 12, 549–557. [Google Scholar]
- Pramanik, S.; Sulistio, Y.A.; Heese, K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol. Neurobiol. 2017, 54, 7401–7459. [Google Scholar] [PubMed]
- Cronin, O.; O’Sullivan, O.; Barton, W.; Cotter, P.D.; Molloy, M.G.; Shanahan, F. Gut microbiota: Implications for sports and exercise medicine. Br. J. Sports Med. 2017, 51, 700–701. [Google Scholar] [CrossRef] [PubMed]
- Hamasaki, H. Exercise and gut microbiota: Clinical implications for the feasibility of Tai Chi. J. Integr. Med. 2017, 15, 270–281. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, C.M.; Davy, B.M.; Hulver, M.W.; Neilson, A.P.; Bennett, B.J.; Davy, K.P. Does Exercise Alter Gut Microbial Composition? A Systematic Review. Med. Sci. Sports Exerc. 2019, 51, 160–167. [Google Scholar] [CrossRef]
- Allen, J.M.; Berg Miller, M.E.; Pence, B.D.; Whitlock, K.; Nehra, V.; Gaskins, H.R.; White, B.A.; Fryer, J.D.; Woods, J.A. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. J. Appl. Physiol. 2015, 118, 1059–1066. [Google Scholar] [CrossRef]
- Evans, J.M.; Morris, L.S.; Marchesi, J.R. The gut microbiome: The role of a virtual organ in the endocrinology of the host. J. Endocrinol. 2013, 218, R37–R47. [Google Scholar] [CrossRef]
- O’Brien, M.T.; O’Sullivan, O.; Claesson, M.J.; Cotter, P.D. The Athlete Gut Microbiome and its Relevance to Health and Performance: A Review. Sports Med. 2022, 52, 119–128. [Google Scholar] [CrossRef]
- Chen, H.; Shen, L.; Liu, Y.; Ma, X.; Long, L.; Ma, X.; Ma, L.; Chen, Z.; Lin, X.; Si, L.; et al. Strength Exercise Confers Protection in Central Nervous System Autoimmunity by Altering the Gut Microbiota. Front. Immunol. 2021, 12, 628629. [Google Scholar] [CrossRef]
- Zuhl, M.; Dokladny, K.; Mermier, C.; Schneider, S.; Salgado, R.; Moseley, P. The effects of acute oral glutamine supplementation on exercise-induced gastrointestinal permeability and heat shock protein expression in peripheral blood mononuclear cells. Cell Stress. Chaperones 2015, 20, 85–93. [Google Scholar] [CrossRef]
- Lambert, G.P. Intestinal barrier dysfunction, endotoxemia, and gastrointestinal symptoms: The ‘canary in the coal mine’ during exercise-heat stress? Med. Sport Sci. 2008, 53, 61–73. [Google Scholar]
- Dokladny, K.; Zuhl, M.N.; Moseley, P.L. Intestinal epithelial barrier function and tight junction proteins with heat and exercise. J. Appl. Physiol. 2016, 120, 692–701. [Google Scholar] [CrossRef] [PubMed]
- Téglás, T.; Ábrahám, D.; Jókai, M.; Kondo, S.; Mohammadi, R.; Fehér, J.; Szabó, D.; Wilhelm, M.; Radák, Z. Exercise combined with a probiotics treatment alters the microbiome, but moderately affects signalling pathways in the liver of male APP/PS1 transgenic mice. Biogerontology 2020, 21, 807–815. [Google Scholar] [CrossRef] [PubMed]
Types of Diseases | Main Symptoms | Model | Gut Microbiota Alterations | Intervention Strategies | References |
---|---|---|---|---|---|
AD | Cognitive impairment, neuropsychiatric symptoms, behavioral disorders | Adults | ↑ Bacteroidetes, Bacteroidaceae, Bacteroides, Rikenellaceae, Alistipes, Blautia, Bilophila ↓ Actinobacteria, Firmicutes, Bifidobacteriaceae, Bifidobacterium, Ruminococcaceae, Turicibacteraceae, Clostridiaceae, Dialister, Turicibacter, Adlercreutzia | Bifidobacterium (Microecologics) | [20] |
↑ Proteobacteria, Gammaproteobacteria, Enterobacteriales, Enterobacteriaceae ↓ Firmicutes, Clostridiaceae, Lachnospiraceae, Ruminococcaceae, Ruminococcus, Blautia | Supplementing the flora that produces short-chain fatty acids | [21] | |||
Mice | ↑ Clostridium pasteurianum, Clostridium innocuum, Clostridium beijerinckii, Firmicutes ↓ Bacteroides ovatus, Bacteroides dorei, Bacteroides vulgatus, Bacteroidetes | FMT | [22] | ||
↑ Bacteroidales ↓ Akkermansia, Parabacteroides | Bacteroides fragilis (Microecologics); FMT | [23] | |||
PD | Cognitive impairment, gait disorder | Adults | ↑ Bifidobacterium dentium, Actinomyces oris, Streptococcus mutans, Lactobacillus fermentum, Escherichia coli, Klebsiella spp., Clostridium leptum, Enterococcus faecium ↓ Roseburia intestinalis, Blautia wexlerae, Faecalibacterium prausnitzii, Eubacterium rectale, Prevotella copri, Roseburia, Eubacterium, Ruminococcus | / | [24] |
↑ Ralstonia, Oxalobacteraceae, Akkermansia, Bacteroides, Oscillospira, Proteobacteria, Verrucomicrobia ↓ Faecalibacterium, Dorea, Coprobacillaceae, Blautia, Coprococcus, Roseburia, Firmicutes | Supplementing butyrice-producing bacteria | [25] | |||
Mice | ↑ Phylum Proteobacteria, Order Turicibacterales, Order Enterobacteriales ↓ Phylum Firmicutes, Order Clostridiales. | FMT | [26] | ||
↓ Verrucomicrobiae | FMT | [27] | |||
ASD | Social communication deficits, restricted interests/repetitive behaviors | Children | ↑ Lactobacillaceae, Bifidobacteraceae, Veillonellaceae, Erysipelotrichaceae, Enterococcaceae, Desulfovibrionaceae, Lactobacillus, Bifidobacterium, Megasphaera, Mitsuokella, Klebsiella ↓ Prevotellaceae, Prevotella, Faecalibacterium, Roseburia | Lactobacillus (Microecologics) | [28] |
Mice | ↑ Lachnospiraceae, Akkermansia, Sutterella ↓ Bacteroides, Parabacteroides, Bacteroidetes | Taurine and 5AV | [29] | ||
MS | Depression, fatigue, pain, cognitive impairment | Adults | ↑ Pseudomonas, Mycoplasma, Blautia, Dorea, Pedobacter ↓ Parabacteroides, Adlercreutzia, Collinsella, Lactobacillus | / | [30] |
Adults | ↑ Euryarchaeota, Verrucomicrobia, Methanobrevibacter, Akkermansia ↓ Butyricimonas, Collinsella, Slackia, Prevotella | / | [31] | ||
ALS | Muscle weakness, atrophy, fasciculations, respiratory dysfunction | Adults | ↑ Bifidobacterium | FMT | [32] |
Adults | ↑ Escherichia coli, Enterobacteriaceae ↓ C. baratii, C. hystoliticum, C. butyricum, C. prefringens, C. botulinum, C. tetan | / | [33] | ||
HD | Chorea, dystonia, cognitive decline | Adults | ↓ Firmicutes, Verrucomicrobia, Akkermansiaceae, Lachnospiraceae, Acidaminococcaceae, Bacteroidaceae, Bifidobacteriaceae, Christensenellaceae, Clostridiaceae, Coriobacteriaceae, Enterobacteriaceae | / | [34] |
Mice | ↑ Bacteroidales, Lactobacillales, Coriobacteriales, Erysipelotrichales, Bacteroidales, Burkholderiale ↓ Clostridiales, Clostridiales | Bacillus subtilis (Microecologics); FMT | [35] | ||
Mice | ↑ Bacteroidetes, Actinobacteria, Proteobacteria ↓ Firmicutes, Deferribacteres | FMT | [36] | ||
ADHD | Inattention, hyperactivity, impulsivity | Children | ↑ Proteobacteria, Shigella ↓ Firmicutes, Bacteroidota | Bifidobacterium bifidum (Microecologics) | [37] |
Children | ↑ Bacteroides uniformis, Bacteroides ovatus, Sutterella stercoricanis, Fusobacteria ↓ Bacteroides coprocola | / | [38] |
Types of Microecologics | Study Model | Doses | Intervention Duration | Results | References |
---|---|---|---|---|---|
Probiotics | |||||
Lactobacillus plantarum PS128 | ASD children | Take one capsule in total (3 × 1010 CFU/capsule) | 4 weeks | Improved oppositional/defiant behavior | [132] |
Probiotic combination (Lactobacillus reuteri, Lactobacillus rhamnosus and Bifidobacterium infantis) | AD mouse model | 1 × 1010 CFU/d | 10 weeks | Improved the spatial memory of rats, and reduced Aβ plaques and inflammation | [133] |
LBS | MS adults | 3.6 × 1012 CFU/d | 2 months | Improved the structure of intestinal flora and inhibited the growth of harmful bacteria | [134] |
Lactobacillus casei Shirota | PD adults | 6.5 × 109 CFU/d | 6 weeks | Improved fecal consistency and reduced abdominal distension and pain | [135] |
Probiotic capsules (Lactobacillus acidophilus, Lactobacillus reuteri, Lactobacillus fermentum and Bifidobacterium) | PD adults | 8 × 109 CFU/d | 12 weeks | Improved cognitive function | [136] |
SLAB51 (Lactobacilli and Bifidobacterium) | AD mouse model | 2 × 1011 CFU/kg/d | 4 months | Improved cognitive impairment | [137] |
Lactobacillus rhamnosus GG ATCC53103 | ADHD children | 8 × 109 CFU/d | 3 months | Improved emotional, physical, social, and academic functioning | [138] |
Akkermansia muciniphila | ALS mouse model | / | 3 months | Alleviated the motor symptoms of ALS mice and prolonged their survival time | [139] |
Prebiotics | |||||
Lactulose | AD mouse model | 200 mg/kg/d | 4 weeks | Improved cognitive impairments | [140] |
MOS | AD adults | (0.12% w/v in the drinking water, with a purity of 85%) replaced twice a week | 8 weeks | Improved behavioral and cognitive disorders | [10] |
Fructooligosaccharides from Morinda officinalis | AD mouse model | 100 mg/kg/d | 8 weeks | Improved learning and memory ability | [141] |
B-GOS® | ASD children | / | 6 weeks | Improved anti-social behavior | [142] |
Synbiotics | |||||
Synbiotic 2000 Forte (Pediococcus pentoseceus 5–33:3, Leuconostoc mesenteroides 32–77:1, L. paracasei ssp. paracasei 19, and L. plantarum 2362, as well as 2.5 g inulin, oat bran, pectin, and resistant starch) | ADHD children | 1 × 1010 CFU/d | 9 weeks | Improved emotion regulation ability | [138] |
Novel synbiotic (PM + LGG) | PD adults | PM (30 mg/kg/d) + LGG (1.5 billion CFU/kg/d) | 5 weeks | Promoted neuroprotection | [143] |
Postbiotics | |||||
Sodium butyrate | PD mouse model | 200 mg/kg/d | 3 weeks | Improved cognitive behavior and coordination ability | [144] |
Sodium butyrate | ALS mouse model | 2% sodium butyrate/d | 2.5 months | Delayed disease onset and prolonged the lifespan of ALS mice | [145] |
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Peng, Z.; Hou, T.; Yang, K.; Zhang, J.; Mao, Y.-H.; Hou, X. Microecologics and Exercise: Targeting the Microbiota–Gut–Brain Axis for Central Nervous System Disease Intervention. Nutrients 2025, 17, 1769. https://doi.org/10.3390/nu17111769
Peng Z, Hou T, Yang K, Zhang J, Mao Y-H, Hou X. Microecologics and Exercise: Targeting the Microbiota–Gut–Brain Axis for Central Nervous System Disease Intervention. Nutrients. 2025; 17(11):1769. https://doi.org/10.3390/nu17111769
Chicago/Turabian StylePeng, Zhixing, Tingting Hou, Keer Yang, Jiangyu Zhang, Yu-Heng Mao, and Xiaohui Hou. 2025. "Microecologics and Exercise: Targeting the Microbiota–Gut–Brain Axis for Central Nervous System Disease Intervention" Nutrients 17, no. 11: 1769. https://doi.org/10.3390/nu17111769
APA StylePeng, Z., Hou, T., Yang, K., Zhang, J., Mao, Y.-H., & Hou, X. (2025). Microecologics and Exercise: Targeting the Microbiota–Gut–Brain Axis for Central Nervous System Disease Intervention. Nutrients, 17(11), 1769. https://doi.org/10.3390/nu17111769