Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes
Abstract
:1. Introduction
2. Role of the Intestinal Microbiota in Inflammation and PD Pathogenesis
3. Role of the Intestinal Microbiota in Metabolism
3.1. SCFAs
3.2. Glucose and Lipid Metabolism
3.3. Vitamins
4. Role of the Intestinal Microbiota in Atypical Parkinsonian Syndromes
5. Role of the Intestinal Microbiota in Future Diagnostic Processes and Treatments
6. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Przewodowska, D.; Marzec, W.; Madetko, N. Novel Therapies for Parkinsonian Syndromes-Recent Progress and Future Perspectives. Front. Mol. Neurosci. 2021, 14, 720220. [Google Scholar] [CrossRef]
- Deutschländer, A.B.; Ross, O.A.; Dickson, D.W.; Wszolek, Z.K. Atypical parkinsonian syndromes: A general neurologist’s perspective. Eur. J. Neurol. 2018, 25, 41–58. [Google Scholar] [CrossRef]
- Khan, I.; De Jesus, O. Frontotemporal Lobe Dementia. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2025. [Google Scholar]
- McFarland, N.R.; Hess, C.W. Recognizing Atypical Parkinsonisms: “Red Flags” and Therapeutic Approaches. Semin. Neurol. 2017, 37, 215–227. [Google Scholar] [PubMed]
- Wenning, G.K.; Stankovic, I.; Vignatelli, L.; Fanciulli, A.; Calandra-Buonaura, G.; Seppi, K.; Palma, J.A.; Meissner, W.G.; Krismer, F.; Berg, D.; et al. The Movement Disorder Society Criteria for the Diagnosis of Multiple System Atrophy. Mov. Disord. 2022, 37, 1131–1148. [Google Scholar] [CrossRef] [PubMed]
- Rana, A.Q.; Qureshi, A.R.; Siddiqui, O.; Sarfraz, Z.; Rana, R.; Shtilbans, A. Prevalence of pain in atypical parkinsonism: A systematic review and meta-analysis. J. Neurol. 2019, 266, 2093–2102. [Google Scholar] [CrossRef]
- Alster, P.; Migda, B.; Madetko-Alster, N. The Significance of High-Density Lipoprotein-Derived Inflammatory Parameters in Atypical Parkinsonisms—Pilot Study. J. Clin. Med. 2025, 14, 2212. [Google Scholar] [CrossRef]
- Lo, R.Y. Epidemiology of atypical parkinsonian syndromes. Tzu Chi Med. J. 2022, 34, 169–181. [Google Scholar] [CrossRef]
- Su, D.; Cui, Y.; He, C.; Yin, P.; Bai, R.; Zhu, J.; Lam, J.S.T.; Zhang, J.; Yan, R.; Zheng, X.; et al. Projections for prevalence of Parkinson’s disease and its driving factors in 195 countries and territories to 2050: Modelling study of Global Burden of Disease Study 2021. BMJ 2025, 388, e080952. [Google Scholar] [CrossRef] [PubMed]
- Lamptey, R.N.L.; Chaulagain, B.; Trivedi, R.; Gothwal, A.; Layek, B.; Singh, J. A Review of the Common Neurodegenerative Disorders: Current Therapeutic Approaches and the Potential Role of Nanotherapeutics. Int. J. Mol. Sci. 2022, 23, 1851. [Google Scholar] [CrossRef]
- Koziorowski, D.; Figura, M.; Milanowski, Ł.M.; Szlufik, S.; Alster, P.; Madetko, N.; Friedman, A. Mechanisms of Neurodegeneration in Various Forms of Parkinsonism-Similarities and Differences. Cells 2021, 10, 656. [Google Scholar] [CrossRef]
- Rietdijk, C.D.; Perez-Pardo, P.; Garssen, J.; van Wezel, R.J.; Kraneveld, A.D. Exploring Braak’s Hypothesis of Parkinson’s Disease. Front. Neurol. 2017, 8, 37. [Google Scholar] [CrossRef] [PubMed]
- Horsager, J.; Andersen, K.B.; Knudsen, K.; Skjærbæk, C.; Fedorova, T.D.; Okkels, N.; Schaeffer, E.; Bonkat, S.K.; Geday, J.; Otto, M.; et al. Brain-first versus body-first Parkinson’s disease: A multimodal imaging case-control study. Brain 2020, 143, 3077–3088. [Google Scholar] [CrossRef] [PubMed]
- Horsager, J.; Knudsen, K.; Sommerauer, M. Clinical and imaging evidence of brain-first and body-first Parkinson’s disease. Neurobiol. Dis. 2022, 164, 105626. [Google Scholar] [CrossRef]
- Rauf, A.; Badoni, H.; Abu-Izneid, T.; Olatunde, A.; Rahman, M.M.; Painuli, S.; Semwal, P.; Wilairatana, P.; Mubarak, M.S. Neuroinflammatory Markers: Key Indicators in the Pathology of Neurodegenerative Diseases. Molecules 2022, 27, 3194. [Google Scholar] [CrossRef]
- Kalyan, M.; Tousif, A.H.; Sonali, S.; Vichitra, C.; Sunanda, T.; Praveenraj, S.S.; Ray, B.; Gorantla, V.R.; Rungratanawanich, W.; Mahalakshmi, A.M.; et al. Role of Endogenous Lipopolysaccharides in Neurological Disorders. Cells 2022, 11, 4038. [Google Scholar] [CrossRef] [PubMed]
- Madetko-Alster, N.; Otto-Ślusarczyk, D.; Wiercińska-Drapało, A.; Koziorowski, D.; Szlufik, S.; Samborska-Ćwik, J.; Struga, M.; Friedman, A.; Alster, P. Clinical Phenotypes of Progressive Supranuclear Palsy-The Differences in Interleukin Patterns. Int. J. Mol. Sci. 2023, 24, 15135. [Google Scholar] [CrossRef]
- Li, F.; Weng, G.; Zhou, H.; Zhang, W.; Deng, B.; Luo, Y.; Tao, X.; Deng, M.; Guo, H.; Zhu, S.; et al. The neutrophil-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, and neutrophil-to-high-density-lipoprotein ratio are correlated with the severity of Parkinson’s disease. Front. Neurol. 2024, 15, 1322228. [Google Scholar] [CrossRef]
- Madetko, N.; Migda, B.; Alster, P.; Turski, P.; Koziorowski, D.; Friedman, A. Platelet-to-lymphocyte ratio and neutrophil-tolymphocyte ratio may reflect differences in PD and MSA-P neuroinflammation patterns. Neurol. Neurochir. Pol. 2022, 56, 148–155. [Google Scholar] [CrossRef]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef]
- Risely, A. Applying the core microbiome to understand host-microbe systems. J. Anim. Ecol. 2020, 89, 1549–1558. [Google Scholar] [CrossRef]
- van de Guchte, M.; Blottière, H.M.; Doré, J. Humans as holobionts: Implications for prevention and therapy. Microbiome 2018, 6, 81. [Google Scholar] [CrossRef] [PubMed]
- Barichella, M.; Severgnini, M.; Cilia, R.; Cassani, E.; Bolliri, C.; Caronni, S.; Ferri, V.; Cancello, R.; Ceccarani, C.; Faierman, S.; et al. Unraveling gut microbiota in Parkinson’s disease and atypical parkinsonism. Mov. Disord. 2019, 34, 396–405. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Li, P.; Li, J.; An, Y.; Wang, M.; Zhong, G. The Differences between Luminal Microbiota and Mucosal Microbiota in Mice. J. Microbiol. Biotechnol. 2020, 30, 287–295. [Google Scholar] [CrossRef]
- Missiego-Beltrán, J.; Beltrán-Velasco, A.I. The Role of Microbial Metabolites in the Progression of Neurodegenerative Diseases-Therapeutic Approaches: A Comprehensive Review. Int. J. Mol. Sci. 2024, 25, 10041. [Google Scholar] [CrossRef]
- Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Park. Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
- Zhu, X.; Shen, J.; Feng, S.; Huang, C.; Wang, H.; Huo, F.; Liu, H. Akkermansia muciniphila, which is enriched in the gut microbiota by metformin, improves cognitive function in aged mice by reducing the proinflammatory cytokine interleukin-6. Microbiome 2023, 11, 120. [Google Scholar] [CrossRef]
- Li, Z.; Lu, G.; Li, Z.; Wu, B.; Luo, E.; Qiu, X.; Guo, J.; Xia, Z.; Zheng, C.; Su, Q.; et al. Altered Actinobacteria and Firmicutes Phylum Associated Epitopes in Patients with Parkinson’s Disease. Front. Immunol. 2021, 12, 632482. [Google Scholar] [CrossRef] [PubMed]
- Yao, Y.; Han, W. Proline Metabolism in Neurological and Psychiatric Disorders. Mol. Cells 2022, 45, 781–788. [Google Scholar] [CrossRef]
- Zhang, K.; Paul, K.C.; Jacobs, J.P.; Chou, H.L.; Duarte Folle, A.; Del Rosario, I.; Yu, Y.; Bronstein, J.M.; Keener, A.M.; Ritz, B. Parkinson’s Disease and the Gut Microbiome in Rural California. J. Parkinsons Dis. 2022, 12, 2441–2452. [Google Scholar] [CrossRef]
- Zuo, S.; Wang, H.; Zhao, Q.; Tang, J.; Wang, M.; Zhang, Y.; Sang, M.; Tian, J.; Wang, P. High levels of Bifidobacteriaceae are associated with the pathogenesis of Parkinson’s disease. Front. Integr. Neurosci. 2022, 16, 1054627. [Google Scholar] [CrossRef]
- Hokari, R.; Tomioka, A. The role of lymphatics in intestinal inflammation. Inflamm. Regen. 2021, 41, 25. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.M.; Islam, F.; -Or-Rashid, M.H.; Mamun, A.A.; Rahaman, M.S.; Islam, M.M.; Meem, A.F.K.; Sutradhar, P.R.; Mitra, S.; Mimi, A.A.; et al. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front. Cell Infect. Microbiol. 2022, 12, 903570. [Google Scholar] [CrossRef]
- Barnhart, M.M.; Chapman, M.R. Curli biogenesis and function. Annu. Rev. Microbiol. 2006, 60, 131–147. [Google Scholar] [CrossRef]
- Sjöbring, U.; Pohl, G.; Olsén, A. Plasminogen, absorbed by Escherichia coli expressing curli or by Salmonella enteritidis expressing thin aggregative fimbriae, can be activated by simultaneously captured tissue-type plasminogen activator (t-PA). Mol. Microbiol. 1994, 14, 443–452. [Google Scholar] [CrossRef]
- Castellino, F.J.; Ploplis, V.A. Structure and function of the plasminogen/plasmin system. Thromb. Haemost. 2005, 93, 647–654. [Google Scholar] [PubMed]
- Chen, S.G.; Stribinskis, V.; Rane, M.J.; Demuth, D.R.; Gozal, E.; Roberts, A.M.; Jagadapillai, R.; Liu, R.; Choe, K.; Shivakumar, B.; et al. Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis elegans. Sci. Rep. 2016, 6, 34477. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.A.; Khan, F.; Song, M. Biofilm-Associated Amyloid Proteins Linked with the Progression of Neurodegenerative Diseases. Int. J. Mol. Sci. 2025, 26, 2695. [Google Scholar] [CrossRef]
- DeWolf, S.E.; Kasimsetty, S.G.; Hawkes, A.A.; Stocks, L.M.; Kurian, S.M.; McKay, D.B. DAMPs Released from Injured Renal Tubular Epithelial Cells Activate Innate Immune Signals in Healthy Renal Tubular Epithelial Cells. Transplantation 2022, 106, 1589–1599. [Google Scholar] [CrossRef]
- Shannon, K.M. Gut-Derived Sterile Inflammation and Parkinson’s Disease. Front. Neurol. 2022, 13, 831090. [Google Scholar] [CrossRef]
- Singh, V.; Lee, G.; Son, H.; Koh, H.; Kim, E.S.; Unno, T.; Shin, J.H. Butyrate producers, “The Sentinel of Gut”: Their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics. Front. Microbiol. 2022, 13, 1103836. [Google Scholar] [CrossRef]
- Rodríguez-Leyva, I.; Chi-Ahumada, E.G.; Carrizales, J.; Rodríguez-Violante, M.; Velázquez-Osuna, S.; Medina-Mier, V.; Martel-Gallegos, M.G.; Zarazúa, S.; Enríquez-Macías, L.; Castro, A.; et al. Parkinson disease and progressive supranuclear palsy: Protein expression in skin. Ann. Clin. Transl. Neurol. 2016, 3, 191–199. [Google Scholar] [CrossRef]
- Sun, Y.; Zhang, M.; Chen, C.C.; Gillilland, M., 3rd; Sun, X.; El-Zaatari, M.; Huffnagle, G.B.; Young, V.B.; Zhang, J.; Hong, S.C.; et al. Stress-induced corticotropin-releasing hormone-mediated NLRP6 inflammasome inhibition and transmissible enteritis in mice. Gastroenterology 2013, 144, 1478–1487.e8. [Google Scholar] [CrossRef]
- Li, Y.; Chen, Y.; Jiang, L.; Zhang, J.; Tong, X.; Chen, D.; Le, W. Intestinal Inflammation and Parkinson’s Disease. Aging Dis. 2021, 12, 2052–2068. [Google Scholar] [CrossRef]
- Nie, S.; Jing, Z.; Wang, J.; Deng, Y.; Zhang, Y.; Ye, Z.; Ge, Y. The link between increased Desulfovibrio and disease severity in Parkinson’s disease. Appl. Microbiol. Biotechnol. 2023, 107, 3033–3045. [Google Scholar] [CrossRef]
- Walker, A.; Czyz, D.M. Oh my gut! Is the microbial origin of neurodegenerative diseases real? Infect. Immun. 2023, 91, e0043722. [Google Scholar] [CrossRef] [PubMed]
- Huynh, V.A.; Takala, T.M.; Murros, K.E.; Diwedi, B.; Saris, P.E.J. Desulfovibrio bacteria enhance alpha-synuclein aggregation in a Caenorhabditis elegans model of Parkinson’s disease. Front. Cell. Infect. Microbiol. 2023, 13, 1181315. [Google Scholar] [CrossRef] [PubMed]
- Andersen, K.K.; Vad, B.S.; Kjaer, L.; Tolker-Nielsen, T.; Christiansen, G.; Otzen, D.E. Pseudomonas aeruginosa rhamnolipid induces fibrillation of human α-synuclein and modulates its effect on biofilm formation. FEBS Lett. 2018, 592, 1484–1496. [Google Scholar] [CrossRef] [PubMed]
- Bleffert, F.; Granzin, J.; Gohlke, H.; Batra-Safferling, R.; Jaeger, K.E.; Kovacic, F. Pseudomonas aeruginosa esterase PA2949, a bacterial homolog of the human membrane esterase ABHD6: Expression, purification and crystallization. Acta Crystallogr. F Struct. Biol. Commun. 2019, 75 Pt 4, 270–277. [Google Scholar] [CrossRef]
- Han, Q.W.; Yuan, Y.H.; Chen, N.H. The therapeutic role of cannabinoid receptors and its agonists or antagonists in Parkinson’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 96, 109745. [Google Scholar] [CrossRef]
- Romo-Vaquero, M.; Fernández-Villalba, E.; Gil-Martinez, A.L.; Cuenca-Bermejo, L.; Espín, J.C.; Herrero, M.T.; Selma, M.V. Urolithins: Potential biomarkers of gut dysbiosis and disease stage in Parkinson’s patients. Food Funct. 2022, 13, 6306–6316. [Google Scholar] [CrossRef]
- Sun, C.Y.; Li, J.R.; Wang, Y.Y.; Lin, S.Y.; Ou, Y.C.; Lin, C.J.; Wang, J.D.; Liao, S.L.; Chen, C.J. p-Cresol Sulfate Caused Behavior Disorders and Neurodegeneration in Mice with Unilateral Nephrectomy Involving Oxidative Stress and Neuroinflammation. Int. J. Mol. Sci. 2020, 21, 6687. [Google Scholar] [CrossRef]
- Singh, Y.; Trautwein, C.; Romani, J.; Salker, M.S.; Neckel, P.H.; Fraccaroli, I.; Abeditashi, M.; Woerner, N.; Admard, J.; Dhariwal, A.; et al. Overexpression of human alpha-Synuclein leads to dysregulated microbiome/metabolites with ageing in a rat model of Parkinson disease. Mol. Neurodegener. 2023, 18, 44. [Google Scholar] [CrossRef]
- Ashurst, J.V.; Nappe, T.M. Isopropanol Toxicity. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2025. [Google Scholar]
- Mathew, A.R.; Di Matteo, G.; La Rosa, P.; Barbati, S.A.; Mannina, L.; Moreno, S.; Tata, A.M.; Cavallucci, V.; Fidaleo, M. Vitamin B12 Deficiency and the Nervous System: Beyond Metabolic Decompensation—Comparing Biological Models and Gaining New Insights into Molecular and Cellular Mechanisms. Int. J. Mol. Sci. 2024, 25, 590. [Google Scholar] [CrossRef]
- Wang, C.; Yang, M.; Liu, D.; Zheng, C. Metabolic rescue of α-synuclein-induced neurodegeneration through propionate supplementation and intestine-neuron signaling in C. elegans. Cell Rep. 2024, 43, 113865. [Google Scholar] [CrossRef] [PubMed]
- Proano, A.C.; Viteri, J.A.; Orozco, E.N.; Calle, M.A.; Costa, S.C.; Reyes, D.V.; German-Montenegro, M.; Moncayo, D.F.; Tobar, A.C.; Moncayo, J.A. Gut Microbiota and Its Repercussion in Parkinson’s Disease: A Systematic Review in Occidental Patients. Neurol. Int. 2023, 15, 750–763. [Google Scholar] [CrossRef]
- Xie, J.; Li, L.F.; Dai, T.Y.; Qi, X.; Wang, Y.; Zheng, T.Z.; Gao, X.Y.; Zhang, Y.J.; Ai, Y.; Ma, L.; et al. Short-Chain Fatty Acids Produced by Ruminococcaceae Mediate α-Linolenic Acid Promote Intestinal Stem Cells Proliferation. Mol. Nutr. Food Res. 2022, 66, e2100408. [Google Scholar] [CrossRef] [PubMed]
- Yang, Q.; Wei, C.; Guo, S.; Liu, J.; Tao, Y. Cloning and characterization of a L-lactate dehydrogenase gene from Ruminococcaceae bacterium CPB6. World J. Microbiol. Biotechnol. 2020, 36, 182. [Google Scholar] [CrossRef] [PubMed]
- Teng, Y.; Mu, J.; Xu, F.; Zhang, X.; Sriwastva, M.K.; Liu, Q.M.; Li, X.; Lei, C.; Sundaram, K.; Hu, X.; et al. Gut bacterial isoamylamine promotes age-related cognitive dysfunction by promoting microglial cell death. Cell Host Microbe 2022, 30, 944–960.e8. [Google Scholar] [CrossRef]
- Tao, Y.; Zhu, X.; Wang, H.; Wang, Y.; Li, X.; Jin, H.; Rui, J. Complete genome sequence of Ruminococcaceae bacterium CPB6: A newly isolated culture for efficient n-caproic acid production from lactate. J. Biotechnol. 2017, 259, 91–94. [Google Scholar] [CrossRef]
- Zhu, Y.; Li, Y.; Zhang, Q.; Song, Y.; Wang, L.; Zhu, Z. Interactions Between Intestinal Microbiota and Neural Mitochondria: A New Perspective on Communicating Pathway from Gut to Brain. Front. Microbiol. 2022, 13, 798917. [Google Scholar] [CrossRef]
- Wei, Y.; Gao, J.; Kou, Y.; Liu, M.; Meng, L.; Zheng, X.; Xu, S.; Liang, M.; Sun, H.; Liu, Z.; et al. The intestinal microbial metabolite desaminotyrosine is an anti-inflammatory molecule that modulates local and systemic immune homeostasis. FASEB J. 2020, 34, 16117–16128. [Google Scholar] [CrossRef] [PubMed]
- Palacios, N.; Hannoun, A.; Flahive, J.; Ward, D.; Goostrey, K.; Deb, A.; Smith, K.M. Effect of Levodopa Initiation on the Gut Microbiota in Parkinson’s Disease. Front. Neurol. 2021, 12, 574529. [Google Scholar] [CrossRef] [PubMed]
- Chu, C.; Yu, L.; Li, Y.; Guo, H.; Zhai, Q.; Chen, W.; Tian, F. Lactobacillus plantarum CCFM405 against Rotenone-Induced Parkinson’s Disease Mice via Regulating Gut Microbiota and Branched-Chain Amino Acids Biosynthesis. Nutrients 2023, 15, 1737. [Google Scholar] [CrossRef]
- Lee, Y.Z.; Cheng, S.H.; Chang, M.Y.; Lin, Y.F.; Wu, C.C.; Tsai, Y.C. Neuroprotective Effects of Lactobacillus plantarum PS128 in a Mouse Model of Parkinson’s Disease: The Role of Gut Microbiota and MicroRNAs. Int. J. Mol. Sci. 2023, 24, 6794. [Google Scholar] [CrossRef]
- Rosario, D.; Boren, J.; Uhlen, M.; Proctor, G.; Aarsland, D.; Mardinoglu, A.; Shoaie, S. Systems Biology Approaches to Understand the Host-Microbiome Interactions in Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 716. [Google Scholar] [CrossRef] [PubMed]
- Andoh, A. Physiological Role of Gut Microbiota for Maintaining Human Health. Digestion 2016, 93, 176–181. [Google Scholar] [CrossRef]
- Flint, H.J. Gut microbial metabolites in health and disease. Gut Microbes 2016, 7, 187–188. [Google Scholar] [CrossRef]
- 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]
- Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and functional importance in the gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
- Yao, Y.; Cai, X.; Fei, W.; Ye, Y.; Zhao, M.; Zheng, C. The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit. Rev. Food Sci. Nutr. 2022, 62, 1–12. [Google Scholar] [CrossRef]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Bermudez-Martin, P.; Becker, J.A.J.; Caramello, N.; Fernandez, S.P.; Costa-Campos, R.; Canaguier, J.; Barbosa, S.; Martinez-Gili, L.; Myridakis, A.; Dumas, M.E.; et al. The microbial metabolite p-Cresol induces autistic-like behaviors in mice by remodeling the gut microbiota. Microbiome 2021, 9, 157. [Google Scholar] [CrossRef] [PubMed]
- O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef]
- Kigerl, K.A.; Ankeny, D.P.; Garg, S.K.; Wei, P.; Guan, Z.; Lai, W.; McTigue, D.M.; Banerjee, R.; Popovich, P.G. System xc− regulates microglia and macrophage glutamate excitotoxicity in vivo. Exp. Neurol. 2012, 233, 333–341. [Google Scholar] [CrossRef]
- Pavek, P. Pregnane X Receptor (PXR)-Mediated Gene Repression and Cross-Talk of PXR with Other Nuclear Receptors via Coactivator Interactions. Front. Pharmacol. 2016, 7, 456. [Google Scholar] [CrossRef]
- Lavelle, A.; Sokol, H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 223–237. [Google Scholar] [CrossRef]
- Ağagündüz, D.; Yılmaz, B.; Şahin, T.; Güneşliol, B.E.; Ayten, Ş.; Russo, P.; Spano, G.; Rocha, J.M.; Bartkiene, E.; Özogul, F. Dairy Lactic Acid Bacteria and Their Potential Function in Dietetics: The Food-Gut-Health Axis. Foods 2021, 10, 3099. [Google Scholar] [CrossRef] [PubMed]
- Pham, L.; Baiocchi, L.; Kennedy, L.; Sato, K.; Meadows, V.; Meng, F.; Huang, C.K.; Kundu, D.; Zhou, T.; Chen, L.; et al. The interplay between mast cells, pineal gland, and circadian rhythm: Links between histamine, melatonin, and inflammatory mediators. J. Pineal Res. 2021, 70, e12699. [Google Scholar] [CrossRef]
- Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef]
- Agnihotri, N.; Mohajeri, M.H. Involvement of Intestinal Microbiota in Adult Neurogenesis and the Expression of Brain-Derived Neurotrophic Factor. Int. J. Mol. Sci. 2022, 23, 15934. [Google Scholar] [CrossRef] [PubMed]
- Müller, P.; Duderstadt, Y.; Lessmann, V.; Müller, N.G. Lactate and BDNF: Key Mediators of Exercise Induced Neuroplasticity? J. Clin. Med. 2020, 9, 1136. [Google Scholar] [CrossRef]
- Guillemin, G.J. Quinolinic acid, the inescapable neurotoxin. FEBS J. 2012, 279, 1356–1365. [Google Scholar] [CrossRef] [PubMed]
- Kaisar, M.M.M.; Pelgrom, L.R.; van der Ham, A.J.; Yazdanbakhsh, M.; Everts, B. Butyrate Conditions Human Dendritic Cells to Prime Type 1 Regulatory T Cells via both Histone Deacetylase Inhibition and G Protein-Coupled Receptor 109A Signaling. Front. Immunol. 2017, 8, 1429. [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]
- Varela, R.B.; Valvassori, S.S.; Lopes-Borges, J.; Mariot, E.; Dal-Pont, G.C.; Amboni, R.T.; Bianchini, G.; Quevedo, J. Sodium butyrate and mood stabilizers block ouabain-induced hyperlocomotion and increase BDNF, NGF and GDNF levels in brain of Wistar rats. J. Psychiatr. Res. 2015, 61, 114–121. [Google Scholar] [CrossRef] [PubMed]
- Barker, R.A.; Björklund, A.; Gash, D.M.; Whone, A.; Van Laar, A.; Kordower, J.H.; Bankiewicz, K.; Kieburtz, K.; Saarma, M.; Booms, S.; et al. GDNF and Parkinson’s Disease: Where Next? A Summary from a Recent Workshop. J. Park. Dis. 2020, 10, 875–891. [Google Scholar] [CrossRef]
- Selma-Royo, M.; Tarrazó, M.; García-Mantrana, I.; Gómez-Gallego, C.; Salminen, S.; Collado, M.C. Shaping Microbiota During the First 1000 Days of Life. Adv. Exp. Med. Biol. 2019, 1125, 3–24. [Google Scholar]
- Garcia-Mantrana, I.; Selma-Royo, M.; Alcantara, C.; Collado, M.C. Shifts on Gut Microbiota Associated to Mediterranean Diet Adherence and Specific Dietary Intakes on General Adult Population. Front. Microbiol. 2018, 9, 890. [Google Scholar] [CrossRef]
- Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
- Vacca, M.; Celano, G.; Calabrese, F.M.; Portincasa, P.; Gobbetti, M.; De Angelis, M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020, 8, 573. [Google Scholar] [CrossRef] [PubMed]
- Maturana, J.L.; Cárdenas, J.P. Insights on the Evolutionary Genomics of the Blautia Genus: Potential New Species and Genetic Content Among Lineages. Front. Microbiol. 2021, 12, 660920. [Google Scholar] [CrossRef]
- Nie, K.; Ma, K.; Luo, W.; Shen, Z.; Yang, Z.; Xiao, M.; Tong, T.; Yang, Y.; Wang, X. Roseburia intestinalis: A Beneficial Gut Organism from the Discoveries in Genus and Species. Front. Cell. Infect. Microbiol. 2021, 11, 757718. [Google Scholar] [CrossRef] [PubMed]
- Zaplana, T.; Miele, S.; Tolonen, A.C. Lachnospiraceae are emerging industrial biocatalysts and biotherapeutics. Front. Bioeng. Biotechnol. 2023, 11, 1324396. [Google Scholar] [CrossRef]
- Palmnäs-Bédard, M.S.A.; Costabile, G.; Vetrani, C.; Åberg, S.; Hjalmarsson, Y.; Dicksved, J.; Riccardi, G.; Landberg, R. The human gut microbiota and glucose metabolism: A scoping review of key bacteria and the potential role of SCFAs. Am. J. Clin. Nutr. 2022, 116, 862–874. [Google Scholar] [CrossRef] [PubMed]
- Ramakrishna, B.S. Role of the gut microbiota in human nutrition and metabolism. J. Gastroenterol. Hepatol. 2013, 28 (Suppl. 4), 9–17. [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]
- Nysten, J.; Van Dijck, P. Can we microbe-manage our vitamin acquisition for better health? PLoS Pathog. 2023, 19, e1011361. [Google Scholar] [CrossRef]
- Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front. Genet. 2015, 6, 148. [Google Scholar] [CrossRef]
- Hossain, K.S.; Amarasena, S.; Mayengbam, S. B Vitamins and Their Roles in Gut Health. Microorganisms 2022, 10, 1168. [Google Scholar] [CrossRef]
- Peterson, C.T.; Rodionov, D.A.; Osterman, A.L.; Peterson, S.N. B Vitamins and Their Role in Immune Regulation and Cancer. Nutrients 2020, 12, 3380. [Google Scholar] [CrossRef] [PubMed]
- Uebanso, T.; Shimohata, T.; Mawatari, K.; Takahashi, A. Functional Roles of B-Vitamins in the Gut and Gut Microbiome. Mol. Nutr. Food Res. 2020, 64, e2000426. [Google Scholar] [CrossRef]
- Javanmardi, S.; Rezaei Tavabe, K.; Rosentrater, K.A.; Solgi, M.; Bahadori, R. Effects of different levels of vitamin B(6) in tank water on the Nile tilapia (Oreochromis niloticus): Growth performance, blood biochemical parameters, intestine and liver histology, and intestinal enzyme activity. Fish. Physiol. Biochem. 2020, 46, 1909–1920. [Google Scholar] [CrossRef] [PubMed]
- Feng, C.; Yan, J.; Luo, T.; Zhang, H.; Zhang, H.; Yuan, Y.; Chen, Y.; Chen, H. Vitamin B12 ameliorates gut epithelial injury via modulating the HIF-1 pathway and gut microbiota. Cell. Mol. Life Sci. 2024, 81, 397. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.; Zadeh, M.; Mohamadzadeh, M. Vitamin B12 coordinates ileal epithelial cell and microbiota functions to resist Salmonella infection in mice. J. Exp. Med. 2022, 219, e20220057. [Google Scholar] [CrossRef]
- Roth, W.; Mohamadzadeh, M. Vitamin B12 and gut-brain homeostasis in the pathophysiology of ischemic stroke. EBioMedicine 2021, 73, 103676. [Google Scholar] [CrossRef]
- Karl, J.P.; Meydani, M.; Barnett, J.B.; Vanegas, S.M.; Barger, K.; Fu, X.; Goldin, B.; Kane, A.; Rasmussen, H.; Vangay, P.; et al. Fecal concentrations of bacterially derived vitamin K forms are associated with gut microbiota composition but not plasma or fecal cytokine concentrations in healthy adults. Am. J. Clin. Nutr. 2017, 106, 1052–1061. [Google Scholar] [CrossRef]
- Pan, M.H.; Maresz, K.; Lee, P.S.; Wu, J.C.; Ho, C.T.; Popko, J.; Mehta, D.S.; Stohs, S.J.; Badmaev, V. Inhibition of TNF-α, IL-1α, and IL-1β by Pretreatment of Human Monocyte-Derived Macrophages with Menaquinone-7 and Cell Activation with TLR Agonists In Vitro. J. Med. Food 2016, 19, 663–669. [Google Scholar] [CrossRef]
- Dihingia, A.; Ozah, D.; Baruah, P.K.; Kalita, J.; Manna, P. Prophylactic role of vitamin K supplementation on vascular inflammation in type 2 diabetes by regulating the NF-κB/Nrf2 pathway via activating Gla proteins. Food Funct. 2018, 9, 450–462. [Google Scholar] [CrossRef]
- Dai, L.; Mafra, D.; Shiels, P.G.; Hackeng, T.M.; Stenvinkel, P.; Schurgers, L.J. Vitamin K and Hallmarks of Ageing: Focus on Diet and Gut Microbiome. Nutrients 2023, 15, 2727. [Google Scholar] [CrossRef] [PubMed]
- Popa, D.S.; Bigman, G.; Rusu, M.E. The Role of Vitamin K in Humans: Implication in Aging and Age-Associated Diseases. Antioxidants 2021, 10, 566. [Google Scholar] [CrossRef] [PubMed]
- Shiels, P.G.; Painer, J.; Natterson-Horowitz, B.; Johnson, R.J.; Miranda, J.J.; Stenvinkel, P. Manipulating the exposome to enable better ageing. Biochem. J. 2021, 478, 2889–2898. [Google Scholar] [CrossRef] [PubMed]
- Bonakdar, M.; Czuba, L.C.; Han, G.; Zhong, G.; Luong, H.; Isoherranen, N.; Vaishnava, S. Gut commensals expand vitamin A metabolic capacity of the mammalian host. Cell Host Microbe 2022, 30, 1084–1092.e5. [Google Scholar] [CrossRef]
- Bourke, C.D.; Berkley, J.A.; Prendergast, A.J. Immune Dysfunction as a Cause and Consequence of Malnutrition. Trends Immunol. 2016, 37, 386–398. [Google Scholar] [CrossRef]
- Zhu, Y.; Self, W.K.; Holtzman, D.M. An emerging role for the gut microbiome in tauopathy. Neurotherapeutics 2024, 21, e00423. [Google Scholar] [CrossRef] [PubMed]
- Mossad, O.; Batut, B.; Yilmaz, B.; Dokalis, N.; Mezö, C.; Nent, E.; Nabavi, L.S.; Mayer, M.; Maron, F.J.M.; Buescher, J.M.; et al. Gut microbiota drives age-related oxidative stress and mitochondrial damage in microglia via the metabolite N(6)-carboxymethyllysine. Nat. Neurosci. 2022, 25, 295–305. [Google Scholar] [CrossRef]
- Buchon, N.; Broderick, N.A.; Chakrabarti, S.; Lemaitre, B. Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in Drosophila. Genes. Dev. 2009, 23, 2333–2344. [Google Scholar] [CrossRef]
- Lei, H.; Crawford, M.S.; McCole, D.F. JAK-STAT Pathway Regulation of Intestinal Permeability: Pathogenic Roles and Therapeutic Opportunities in Inflammatory Bowel Disease. Pharmaceuticals 2021, 14, 840. [Google Scholar] [CrossRef]
- Zhao, J.; Bang, S.; Furutani, K.; McGinnis, A.; Jiang, C.; Roberts, A.; Donnelly, C.R.; He, Q.; James, M.L.; Berger, M.; et al. PD-L1/PD-1 checkpoint pathway regulates hippocampal neuronal excitability and learning and memory behavior. Neuron 2023, 111, 2709–2726.e9. [Google Scholar] [CrossRef]
- Zhou, Y.; Liu, Z.; Chen, T. Gut Microbiota: A Promising Milestone in Enhancing the Efficacy of PD1/PD-L1 Blockade Therapy. Front. Oncol. 2022, 12, 847350. [Google Scholar] [CrossRef]
- Chen, X.; Firulyova, M.; Manis, M.; Herz, J.; Smirnov, I.; Aladyeva, E.; Wang, C.; Bao, X.; Finn, M.B.; Hu, H.; et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature 2023, 615, 668–677. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, A.M.; Krout, I.N.; White, A.C.; Sampson, T.R. Microbiome-based therapeutics for Parkinson’s disease. Neurotherapeutics 2024, 21, e00462. [Google Scholar] [CrossRef]
- DuPont, H.L.; Suescun, J.; Jiang, Z.D.; Brown, E.L.; Essigmann, H.T.; Alexander, A.S.; DuPont, A.W.; Iqbal, T.; Utay, N.S.; Newmark, M.; et al. Fecal microbiota transplantation in Parkinson’s disease—A randomized repeat-dose, placebo-controlled clinical pilot study. Front. Neurol. 2023, 14, 1104759. [Google Scholar] [CrossRef]
- Cheng, Y.; Tan, G.; Zhu, Q.; Wang, C.; Ruan, G.; Ying, S.; Qie, J.; Hu, X.; Xiao, Z.; Xu, F.; et al. Efficacy of fecal microbiota transplantation in patients with Parkinson’s disease: Clinical trial results from a randomized, placebo-controlled design. Gut Microbes 2023, 15, 2284247. [Google Scholar] [CrossRef]
- Scheperjans, F.; Levo, R.; Bosch, B.; Lääperi, M.; Pereira, P.A.B.; Smolander, O.P.; Aho, V.T.E.; Vetkas, N.; Toivio, L.; Kainulainen, V.; et al. Fecal Microbiota Transplantation for Treatment of Parkinson Disease: A Randomized Clinical Trial. JAMA Neurol. 2024, 81, 925–938. [Google Scholar] [CrossRef] [PubMed]
- Tian, H.; Wang, J.; Feng, R.; Zhang, R.; Liu, H.; Qin, C.; Meng, L.; Chen, Y.; Fu, Y.; Liang, D.; et al. Efficacy of faecal microbiota transplantation in patients with progressive supranuclear palsy-Richardson’s syndrome: A phase 2, single centre, randomised clinical trial. EClinicalMedicine 2023, 58, 101888. [Google Scholar] [CrossRef] [PubMed]
- Forloni, G. Alpha Synuclein: Neurodegeneration and Inflammation. Int. J. Mol. Sci. 2023, 24, 5914. [Google Scholar] [CrossRef] [PubMed]
- Wijeyekoon, R.S.; Kronenberg-Versteeg, D.; Scott, K.M.; Hayat, S.; Kuan, W.L.; Evans, J.R.; Breen, D.P.; Cummins, G.; Jones, J.L.; Clatworthy, M.R.; et al. Peripheral innate immune and bacterial signals relate to clinical heterogeneity in Parkinson’s disease. Brain Behav. Immun. 2020, 87, 473–488. [Google Scholar] [CrossRef]
- Su, Y.; Shi, C.; Wang, T.; Liu, C.; Yang, J.; Zhang, S.; Fan, L.; Zheng, H.; Li, X.; Luo, H.; et al. Dysregulation of peripheral monocytes and pro-inflammation of alpha-synuclein in Parkinson’s disease. J. Neurol. 2022, 269, 6386–6394. [Google Scholar] [CrossRef]
- La Vitola, P.; Balducci, C.; Baroni, M.; Artioli, L.; Santamaria, G.; Castiglioni, M.; Cerovic, M.; Colombo, L.; Caldinelli, L.; Pollegioni, L.; et al. Peripheral inflammation exacerbates α-synuclein toxicity and neuropathology in Parkinson’s models. Neuropathol. Appl. Neurobiol. 2021, 47, 43–60. [Google Scholar] [CrossRef]
- Reddy, K.; Dieriks, B.V. Multiple system atrophy: α-Synuclein strains at the neuron-oligodendrocyte crossroad. Mol. Neurodegener. 2022, 17, 77. [Google Scholar] [CrossRef]
- Holec, S.A.M.; Lee, J.; Oehler, A.; Batia, L.; Wiggins-Gamble, A.; Lau, J.; Ooi, F.K.; Merz, G.E.; Wang, M.; Mordes, D.A.; et al. The E46K mutation modulates α-synuclein prion replication in transgenic mice. PLoS Pathog. 2022, 18, e1010956. [Google Scholar] [CrossRef] [PubMed]
- Pan, L.; Li, C.; Meng, L.; Tian, Y.; He, M.; Yuan, X.; Zhang, G.; Zhang, Z.; Xiong, J.; Chen, G.; et al. Tau accelerates α-synuclein aggregation and spreading in Parkinson’s disease. Brain 2022, 145, 3454–3471. [Google Scholar] [CrossRef]
- Engen, P.A.; Dodiya, H.B.; Naqib, A.; Forsyth, C.B.; Green, S.J.; Voigt, R.M.; Kordower, J.H.; Mutlu, E.A.; Shannon, K.M.; Keshavarzian, A. The Potential Role of Gut-Derived Inflammation in Multiple System Atrophy. J. Park. Dis. 2017, 7, 331–346. [Google Scholar] [CrossRef] [PubMed]
- Tan, A.H.; Chong, C.W.; Song, S.L.; Teh, C.S.J.; Yap, I.K.S.; Loke, M.F.; Tan, Y.Q.; Yong, H.S.; Mahadeva, S.; Lang, A.E.; et al. Altered gut microbiome and metabolome in patients with multiple system atrophy. Mov. Disord. 2018, 33, 174–176. [Google Scholar] [CrossRef] [PubMed]
- Wan, L.; Zhou, X.; Wang, C.; Chen, Z.; Peng, H.; Hou, X.; Peng, Y.; Wang, P.; Li, T.; Yuan, H.; et al. Alterations of the Gut Microbiota in Multiple System Atrophy Patients. Front. Neurosci. 2019, 13, 1102. [Google Scholar] [CrossRef]
- Nagoya University. New Research Shows Link Between Gut Microbiota and Parkinson’s Disease. 2024. Available online: https://www.labmanager.com/new-research-shows-link-between-gut-microbiata-and-parkinson-s-disease-32393 (accessed on 20 January 2025).
- Nishiwaki, H.; Ueyama, J.; Ito, M.; Hamaguchi, T.; Takimoto, K.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Mori, H.; Kurokawa, K.; et al. Meta-analysis of shotgun sequencing of gut microbiota in Parkinson’s disease. NPJ Park. Dis. 2024, 10, 106. [Google Scholar] [CrossRef]
- Zhao, Z.; Chen, J.; Zhao, D.; Chen, B.; Wang, Q.; Li, Y.; Chen, J.; Bai, C.; Guo, X.; Hu, N.; et al. Microbial biomarker discovery in Parkinson’s disease through a network-based approach. NPJ Park. Dis. 2024, 10, 203. [Google Scholar] [CrossRef]
- Munoz-Pinto, M.F.; Candeias, E.; Melo-Marques, I.; Esteves, A.R.; Maranha, A.; Magalhães, J.D.; Carneiro, D.R.; Sant’Anna, M.; Pereira-Santos, A.R.; Abreu, A.E.; et al. Gut-first Parkinson’s disease is encoded by gut dysbiome. Mol. Neurodegener. 2024, 19, 78. [Google Scholar] [CrossRef]
- Borzabadi, S.; Oryan, S.; Eidi, A.; Aghadavod, E.; Daneshvar Kakhaki, R.; Tamtaji, O.R.; Taghizadeh, M.; Asemi, Z. The Effects of Probiotic Supplementation on Gene Expression Related to Inflammation, Insulin and Lipid in Patients with Parkinson’s Disease: A Randomized, Double-blind, PlaceboControlled Trial. Arch. Iran. Med. 2018, 21, 289–295. [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] [PubMed]
- Becker, A.; Schmartz, G.P.; Gröger, L.; Grammes, N.; Galata, V.; Philippeit, H.; Weiland, J.; Ludwig, N.; Meese, E.; Tierling, S.; et al. Effects of Resistant Starch on Symptoms, Fecal Markers, and Gut Microbiota in Parkinson’s Disease—The RESISTA-PD Trial. Genom. Proteom. Bioinform. 2022, 20, 274–287. [Google Scholar] [CrossRef]
- 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] [PubMed]
- 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 Park. Dis. 2022, 8, 62. [Google Scholar] [CrossRef]
- Yang, X.; He, X.; Xu, S.; Zhang, Y.; Mo, C.; Lai, Y.; Song, Y.; Yan, Z.; Ai, P.; Qian, Y.; et al. Effect of Lacticaseibacillus paracasei strain Shirota supplementation on clinical responses and gut microbiome in Parkinson’s disease. Food Funct. 2023, 14, 6828–6839. [Google Scholar] [CrossRef] [PubMed]
- Loeffler, D.A. Influence of Normal Aging on Brain Autophagy: A Complex Scenario. Front. Aging Neurosci. 2019, 11, 49. [Google Scholar] [CrossRef]
- Ahmed, S.; Busetti, A.; Fotiadou, P.; Vincy Jose, N.; Reid, S.; Georgieva, M.; Brown, S.; Dunbar, H.; Beurket-Ascencio, G.; Delday, M.I.; et al. In vitro Characterization of Gut Microbiota-Derived Bacterial Strains with Neuroprotective Properties. Front. Cell Neurosci. 2019, 13, 402. [Google Scholar] [CrossRef]
- Głowacka, P.; Oszajca, K.; Pudlarz, A.; Szemraj, J.; Witusik-Perkowska, M. Postbiotics as Molecules Targeting Cellular Events of Aging Brain-The Role in Pathogenesis, Prophylaxis and Treatment of Neurodegenerative Diseases. Nutrients 2024, 16, 2244. [Google Scholar] [CrossRef]
- Tanner, C. Microbiota Intervention to Change the Response of Parkinson’s Disease. 2019. Available online: https://clinicaltrials.ucsf.edu/trial/NCT03575195 (accessed on 20 January 2025).
- Ho, J. To Assess the Efficacy and Safety of Ceftriaxone in Patients with Mild to Moderate Parkinson’s Disease Dementia. 2019. Available online: https://clinicaltrial.be/nl/details/69615?per_page=100&only_recruiting=0&only_eligible=0&only_active=0 (accessed on 20 January 2025).
- Wang, J.Y.; Doudna, J.A. CRISPR technology: A decade of genome editing is only the beginning. Science 2023, 379, eadd8643. [Google Scholar] [CrossRef]
- Liu, J.; Lv, X.; Ye, T.; Zhao, M.; Chen, Z.; Zhang, Y.; Yang, W.; Xie, H.; Zhan, L.; Chen, L.; et al. Microbiota-microglia crosstalk between Blautia producta and neuroinflammation of Parkinson’s disease: A bench-to-bedside translational approach. Brain Behav. Immun. 2024, 117, 270–282. [Google Scholar] [CrossRef]
Bacteria | Activity | Effect |
---|---|---|
Akkermansia | degrades mucin regulates T regulatory cell activity regulates SCFA production inhibits IL-6 dependent pathway | intestinal passage prolongation cognitive function improvement |
Actinobacteria | increases glutamate level, promoting neuronal excitation forms putrescine and spermidine | enhancement of aggregation of α-synuclein risk of increased oxidative stress |
Desulfovibrionaceae | produces magnetite (Fe3O4) | excessive cytochrome c release in mitochondria iron accumulation and increase in the level of reactive oxygen species production |
Proteobacteria | rhamnolipid (RL) interacts with α-synuclein molecules disables endocannabinoid receptors | risk of neurodegeneration symptoms occurrence |
Prevotellaceae | decreases toxic polyglutamine (polyQ) aggregation | decreased constipation |
Firmicutes (type) Bacilli (class) Lactobacillus Enterococcus Streptococcaceae Clostridium (class) Lachnospiraceae Ruminococcaceae | inhibits isopropanol production ferments pyruvate to propanoate I promotes BCAAs production produces p-cresol decreases circulating BDNF disrupts intercellular connections inhibits potassium channels produces SCFAs produces secondary bile acids produces α-linolenic acid (ALA) | potential protective role against inflammation and cell death (in AD) intestinal microbiota composition modification inflammation reduction depression-like, anxiety-like and cognitive impairment symptoms development increased intestinal permeability increased cellular death and intestinal permeability enteric pathogen inhibition mucosal immunity regulation apoptosis of microglial cells |
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
Przewodowska, D.; Alster, P.; Madetko-Alster, N. Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes. Int. J. Mol. Sci. 2025, 26, 3928. https://doi.org/10.3390/ijms26093928
Przewodowska D, Alster P, Madetko-Alster N. Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes. International Journal of Molecular Sciences. 2025; 26(9):3928. https://doi.org/10.3390/ijms26093928
Chicago/Turabian StylePrzewodowska, Dominika, Piotr Alster, and Natalia Madetko-Alster. 2025. "Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes" International Journal of Molecular Sciences 26, no. 9: 3928. https://doi.org/10.3390/ijms26093928
APA StylePrzewodowska, D., Alster, P., & Madetko-Alster, N. (2025). Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes. International Journal of Molecular Sciences, 26(9), 3928. https://doi.org/10.3390/ijms26093928