Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship
Abstract
:1. Relationship between Gut Microbiota and Brain Function
2. Gut Microbiota and Alzheimer’s Disease
3. How to Study the Relationship between GM and AD
3.1. In Vitro Study
3.2. In Vivo Study
3.3. Human Study
3.4. Relevant Techniques
4. GM-Directed Therapeutic Options to Ameliorate the Progression of AD
4.1. Prebiotics
4.2. Probiotics
4.3. Diet
4.4. Fecal Microbiota Transplantation
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut Microbes and the Brain: Paradigm Shift in Neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hugon, P.; Dufour, J.-C.; Colson, P.; Fournier, P.-E.; Sallah, K.; Raoult, D. A comprehensive repertoire of prokaryotic species identified in human beings. Lancet Infect. Dis. 2015, 15, 1211–1219. [Google Scholar] [CrossRef] [PubMed]
- Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nat. Rev. Genet. 2012, 13, 260–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
- Foster, J.A.; McVey Neufeld, K.-A. Gut–brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef] [PubMed]
- Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef] [Green Version]
- Stolfi, C.; Maresca, C.; Monteleone, G.; Laudisi, F. Implication of Intestinal Barrier Dysfunction in Gut Dysbiosis and Diseases. Biomedicines 2022, 10, 289. [Google Scholar] [CrossRef]
- Lyte, M. Host-microbiota neuroendocrine interactions influencing brain and behavior. Gut Microbes 2014, 5, 381–389. [Google Scholar] [CrossRef] [Green Version]
- Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938. [Google Scholar] [CrossRef] [Green Version]
- Borre, Y.E.; O’Keeffe, G.W.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol. Med. 2014, 20, 509–518. [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] [Green Version]
- Cryan, J.F.; O’Riordan, K.J.; Sandhu, K.; Peterson, V.; Dinan, T.G. The gut microbiome in neurological disorders. Lancet Neurol. 2020, 19, 179–194. [Google Scholar] [CrossRef] [PubMed]
- Tremlett, H.; Bauer, K.C.; Appel-Cresswell, S.; Finlay, B.B.; Waubant, E. The gut microbiome in human neurological disease: A review. Ann. Neurol. 2017, 81, 369–382. [Google Scholar] [CrossRef] [PubMed]
- Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Needham, B.D.; Kaddurah-Daouk, R.; Mazmanian, S.K. Gut microbial molecules in behavioural and neurodegenerative conditions. Nat. Rev. Neurosci. 2020, 21, 717–731. [Google Scholar] [CrossRef]
- Gubert, C.; Kong, G.; Renoir, T.; Hannan, A.J. Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol. Dis. 2020, 134, 104621. [Google Scholar] [CrossRef]
- Conte, C.; Sichetti, M.; Traina, G. Gut–Brain Axis: Focus on Neurodegeneration and Mast Cells. Appl. Sci. 2020, 10, 1828. [Google Scholar] [CrossRef] [Green Version]
- Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [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]
- Hill-Burns, E.M.; Debelius, J.W.; Morton, J.T.; Wissemann, W.T.; Lewis, M.R.; Wallen, Z.D.; Peddada, S.D.; Factor, S.A.; Molho, E.; Zabetian, C.P.; et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov. Disord. 2017, 32, 739–749. [Google Scholar] [CrossRef] [Green Version]
- Felice, V.D.; Quigley, E.M.; Sullivan, A.M.; O’Keeffe, G.W.; O’Mahony, S.M. Microbiota-gut-brain signalling in Parkinson’s disease: Implications for non-motor symptoms. Park. Relat. Disord. 2016, 27, 1–8. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- Kong, G.; Cao, K.-A.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] [PubMed]
- 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]
- Kong, G.; Ellul, S.; Narayana, V.K.; Kanojia, K.; Ha, H.T.T.; Li, S.; Renoir, T.; Cao, K.-A.L.; Hannan, A.J. An integrated metagenomics and metabolomics approach implicates the microbiota-gut-brain axis in the pathogenesis of Huntington’s disease. Neurobiol. Dis. 2021, 148, 105199. [Google Scholar] [CrossRef]
- De-Paula, V.J.; Radanovic, M.; Diniz, B.S.; Forlenza, O.V. Alzheimer’s Disease. Subcell. Biochem. 2012, 65, 329–352. [Google Scholar] [CrossRef]
- Janeiro, M.H.; Ramírez, M.J.; Solas, M. Dysbiosis and Alzheimer’s Disease: Cause or Treatment Opportunity? Cell. Mol. Neurobiol. 2022, 42, 377–387. [Google Scholar] [CrossRef]
- Pistollato, F.; Sumalla Cano, S.; Elio, I.; Masias Vergara, M.; Giampieri, F.; Battino, M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr. Rev. 2016, 74, 624–634. [Google Scholar] [CrossRef] [Green Version]
- Shabbir, U.; Arshad, M.S.; Sameen, A.; Oh, D.-H. Crosstalk between Gut and Brain in Alzheimer’s Disease: The Role of Gut Microbiota Modulation Strategies. Nutrients 2021, 13, 690. [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] [Green Version]
- Bostanciklioğlu, M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J. Appl. Microbiol. 2019, 127, 954–967. [Google Scholar] [CrossRef] [PubMed]
- Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The Gut Microbiota and Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Guo, M.; Peng, J.; Huang, X.; Xiao, L.; Huang, F.; Zuo, Z. Gut Microbiome Features of Chinese Patients Newly Diagnosed with Alzheimer’s Disease or Mild Cognitive Impairment. J. Alzheimer’s Dis. 2021, 80, 299–310. [Google Scholar] [CrossRef] [PubMed]
- Haran, J.P.; Bhattarai, S.K.; Foley, S.E.; Dutta, P.; Ward, D.V.; Bucci, V.; McCormick, B.A. Alzheimer’s Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. Mbio 2019, 10, e00632-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caini, S.; Bagnoli, S.; Palli, D.; Saieva, C.; Ceroti, M.; Bendinelli, B.; Assedi, M.; Masala, G. Total and cancer mortality in a cohort of ulcerative colitis and Crohn’s disease patients: The Florence inflammatory bowel disease study, 1978–2010. Dig. Liver Dis. 2016, 48, 1162–1167. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.-H.; Lin, C.-L.; Kao, C.-H. Irritable Bowel Syndrome Is Associated with an Increased Risk of Dementia: A Nationwide Population-Based Study. PLoS ONE 2016, 11, e0144589. [Google Scholar] [CrossRef]
- Ling, Z.; Zhu, M.; Liu, X.; Shao, L.; Cheng, Y.; Yan, X.; Jiang, R.; Wu, S. Fecal Fungal Dysbiosis in Chinese Patients with Alzheimer’s Disease. Front. Cell Dev. Biol. 2020, 8, 631460. [Google Scholar] [CrossRef]
- Ling, Z.; Zhu, M.; Yan, X.; Cheng, Y.; Shao, L.; Liu, X.; Jiang, R.; Wu, S. Structural and Functional Dysbiosis of Fecal Microbiota in Chinese Patients with Alzheimer’s Disease. Front. Cell Dev. Biol. 2020, 8, 634069. [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]
- Morris, G.; Berk, M.; Carvalho, A.; Caso, J.R.; Sanz, Y.; Walder, K.; Maes, M. The Role of the Microbial Metabolites Including Tryptophan Catabolites and Short Chain Fatty Acids in the Pathophysiology of Immune-Inflammatory and Neuroimmune Disease. Mol. Neurobiol. 2017, 54, 4432–4451. [Google Scholar] [CrossRef]
- Li, B.; He, Y.; Ma, J.; Huang, P.; Du, J.; Cao, L.; Wang, Y.; Xiao, Q.; Tang, H.; Chen, S. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimer’s Dement. 2019, 15, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
- Paranjapye, N.; Daggett, V. De Novo Designed α-Sheet Peptides Inhibit Functional Amyloid Formation of Streptococcus mutans Biofilms. J. Mol. Biol. 2018, 430, 3764–3773. [Google Scholar] [CrossRef] [PubMed]
- Zhan, X.; Stamova, B.; Sharp, F.R. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer’s Disease Brain: A Review. Front. Aging Neurosci. 2018, 10, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hrncir, T.; Hrncirova, L.; Kverka, M.; Tlaskalova-Hogenova, H. The role of gut microbiota in intestinal and liver diseases. Lab. Anim. 2019, 53, 271–280. [Google Scholar] [CrossRef]
- Takashima, A. Tau aggregation is a therapeutic target for Alzheimer’s disease. Curr. Alzheimer Res. 2010, 7, 665–669. [Google Scholar] [CrossRef]
- Vogt, N.M.; Romano, K.A.; Darst, B.F.; Engelman, C.D.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Blennow, K.; Zetterberg, H.; Bendlin, B.B.; et al. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease. Alzheimer’s Res. Ther. 2018, 10, 124. [Google Scholar] [CrossRef] [Green Version]
- Wei, S.; Peng, W.; Mai, Y.; Li, K.; Wei, W.; Hu, L.; Zhu, S.; Zhou, H.; Jie, W.; Wei, Z.; et al. Outer membrane vesicles enhance tau phosphorylation and contribute to cognitive impairment. J. Cell. Physiol. 2020, 235, 4843–4855. [Google Scholar] [CrossRef]
- Kim, M.-S.; Kim, Y.; Choi, H.; Kim, W.; Park, S.; Lee, D.; Kim, D.K.; Kim, H.J.; Choi, H.; Hyun, D.-W.; et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 2020, 69, 283–294. [Google Scholar] [CrossRef]
- Harach, T.; Marungruang, N.; Duthilleul, N.; Cheatham, V.; Mc Coy, K.D.; Frisoni, G.B.; 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] [Green Version]
- Park, A.-M.; Omura, S.; Fujita, M.; Sato, F.; Tsunoda, I. Helicobacter pylori and gut microbiota in multiple sclerosis versus Alzheimer’s disease: 10 pitfalls of microbiome studies. Clin. Exp. Neuroimmunol. 2017, 8, 215–232. [Google Scholar] [CrossRef] [Green Version]
- Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Qureshi, I.A.; Mehler, M.F. Towards a ‘systems’-level understanding of the nervous system and its disorders. Trends Neurosci. 2013, 36, 674–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, J.; Xu, J.; Yang, B.; Chen, K.; Kong, Y.; Fang, N.; Gong, T.; Wang, F.; Ling, Z.; Liu, J. Effect of Clostridium butyricum against Microglia-Mediated Neuroinflammation in Alzheimer’s Disease via Regulating Gut Microbiota and Metabolites Butyrate. Mol. Nutr. Food Res. 2020, 64, e1900636. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Sun, Y.-X.; Jiang, X.-J.; Lu, B.; Gao, Q.; Chen, Y.-F.; Wu, D.-B.; Zeng, W.-Y.; Yang, L.; Li, H.-H.; Yu, B. Roles of Gut Microbiota in Pathogenesis of Alzheimer’s Disease and Therapeutic Effects of Chinese Medicine. Chin. J. Integr. Med. 2022, 28, 1048–1056. [Google Scholar] [CrossRef]
- Rutsch, A.; Kantsjö, J.B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179. [Google Scholar] [CrossRef]
- McCarville, J.L.; Chen, G.Y.; Cuevas, V.D.; Troha, K.; Ayres, J.S. Microbiota Metabolites in Health and Disease. Annu. Rev. Immunol. 2020, 38, 147–170. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Manyevitch, R.; Protas, M.; Scarpiello, S.; DeLiso, M.; Bass, B.; Nanajian, A.; Chang, M.; Thompson, S.M.; Khoury, N.; Gonnella, R.; et al. Evaluation of Metabolic and Synaptic Dysfunction Hypotheses of Alzheimer’s Disease (AD): A Meta-Analysis of CSF Markers. Curr. Alzheimer Res. 2018, 15, 164–181. [Google Scholar] [CrossRef]
- Wu, L.; Han, Y.; Zheng, Z.; Peng, G.; Liu, P.; Yue, S.; Zhu, S.; Chen, J.; Lv, H.; Shao, L.; et al. Altered Gut Microbial Metabolites in Amnestic Mild Cognitive Impairment and Alzheimer’s Disease: Signals in Host–Microbe Interplay. Nutrients 2021, 13, 228. [Google Scholar] [CrossRef]
- Unger, M.M.; Spiegel, J.; Dillmann, K.-U.; Grundmann, D.; Philippeit, H.; Bürmann, J.; Faßbender, K.; Schwiertz, A.; Schäfer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Whiley, L.; on behalf of AddNeuroMed consortium; Chappell, K.E.; D’Hondt, E.; Lewis, M.R.; Jiménez, B.; Snowden, S.G.; Soininen, H.; Kłoszewska, I.; Mecocci, P.; et al. Metabolic phenotyping reveals a reduction in the bioavailability of serotonin and kynurenine pathway metabolites in both the urine and serum of individuals living with Alzheimer’s disease. Alzheimer’s Res. Ther. 2021, 13, 20. [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]
- Parker, A.; Fonseca, S.; Carding, S.R. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2020, 11, 135–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tiso, M.; Schechter, A.N. Nitrate Reduction to Nitrite, Nitric Oxide and Ammonia by Gut Bacteria under Physiological Conditions. PLoS ONE 2015, 10, e0119712. [Google Scholar] [CrossRef] [Green Version]
- Stan, T.L.; Soylu-Kucharz, R.; Burleigh, S.; Prykhodko, O.; Cao, L.; Franke, N.; Sjögren, M.; Haikal, C.; Hållenius, F.; Björkqvist, M. Increased intestinal permeability and gut dysbiosis in the R6/2 mouse model of Huntington’s disease. Sci. Rep. 2020, 10, 18270. [Google Scholar] [CrossRef]
- Beaumont, M.; Andriamihaja, M.; Lan, A.; Khodorova, N.; Audebert, M.; Blouin, J.-M.; Grauso, M.; Lancha, L.; Benetti, P.-H.; Benamouzig, R.; et al. Detrimental effects for colonocytes of an increased exposure to luminal hydrogen sulfide: The adaptive response. Free. Radic. Biol. Med. 2016, 93, 155–164. [Google Scholar] [CrossRef] [PubMed]
- Ohta, S. Molecular hydrogen as a preventive and therapeutic medical gas: Initiation, development and potential of hydrogen medicine. Pharmacol. Ther. 2014, 144, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Eain, M.M.G.; Baginska, J.; Greenhalgh, K.; Fritz, J.V.; Zenhausern, F.; Wilmes, P. Engineering Solutions for Representative Models of the Gastrointestinal Human-Microbe Interface. Engineering 2017, 3, 60–65. [Google Scholar] [CrossRef] [Green Version]
- Sommer, M.O. Advancing gut microbiome research using cultivation. Curr. Opin. Microbiol. 2015, 27, 127–132. [Google Scholar] [CrossRef]
- Ingber, D.E. Reverse Engineering Human Pathophysiology with Organs-on-Chips. Cell 2016, 164, 1105–1109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Q.; Liu, J.; Wang, X.; Feng, L.; Wu, J.; Zhu, X.; Wen, W.; Gong, X. Organ-on-a-chip: Recent breakthroughs and future prospects. Biomed. Eng. Online 2020, 19, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yesil-Celiktas, O.; Hassan, S.; Miri, A.K.; Maharjan, S.; Al-Kharboosh, R.; Quiñones-Hinojosa, A.; Zhang, Y.S. Mimicking Human Pathophysiology in Organ-on-Chip Devices. Adv. Biosyst. 2018, 2, 1800109. [Google Scholar] [CrossRef]
- Ceppa, F.A.; Izzo, L.; Sardelli, L.; Raimondi, I.; Tunesi, M.; Albani, D.; Giordano, C. Human Gut-Microbiota Interaction in Neurodegenerative Disorders and Current Engineered Tools for Its Modeling. Front. Cell. Infect. Microbiol. 2020, 10, 297. [Google Scholar] [CrossRef] [PubMed]
- Kim, R.; Attayek, P.J.; Wang, Y.; Furtado, K.L.; Tamayo, R.; E Sims, C.; Allbritton, N.L. An in vitro intestinal platform with a self-sustaining oxygen gradient to study the human gut/microbiome interface. Biofabrication 2019, 12, 015006. [Google Scholar] [CrossRef] [PubMed]
- Ambrosini, Y.M.; Shin, W.; Min, S.; Kim, H.J. Microphysiological Engineering of Immune Responses in Intestinal Inflammation. Immune Netw. 2020, 20, e13. [Google Scholar] [CrossRef]
- Choi, Y.J.; Chae, S.; Kim, J.H.; Barald, K.F.; Park, J.Y.; Lee, S.-H. Neurotoxic amyloid beta oligomeric assemblies recreated in microfluidic platform with interstitial level of slow flow. Sci. Rep. 2013, 3, srep01921. [Google Scholar] [CrossRef] [Green Version]
- Raimondi, M.T.; Albani, D.; Giordano, C. An Organ-On-A-Chip Engineered Platform to Study the Microbiota–Gut–Brain Axis in Neurodegeneration. Trends Mol. Med. 2019, 25, 737–740. [Google Scholar] [CrossRef] [Green Version]
- Bry, L.; Falk, P.G.; Midtvedt, T.; Gordon, J.I. A Model of Host-Microbial Interactions in an Open Mammalian Ecosystem. Science 1996, 273, 1380–1383. [Google Scholar] [CrossRef]
- Gerard, P.; Beguet, F.; Lepercq, P.; Rigottier-Gois, L.; Rochet, V.; Andrieux, C.; Juste, C. Gnotobiotic rats harboring human intestinal microbiota as a model for studying cholesterol-to-coprostanol conversion. FEMS Microbiol. Ecol. 2004, 47, 337–343. [Google Scholar] [CrossRef] [Green Version]
- Meurens, F.; Berri, M.; Siggers, R.H.; Willing, B.P.; Salmon, H.; Van Kessel, A.G.; Gerdts, V. Commensal Bacteria and Expression of Two Major Intestinal Chemokines, TECK/CCL25 and MEC/CCL28, and Their Receptors. PLoS ONE 2007, 2, e677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rawls, J.F.; Samuel, B.S.; Gordon, J.I. Gnotobiotic zebrafish reveal evolutionarily conserved responses to the gut microbiota. Proc. Natl. Acad. Sci. USA 2004, 101, 4596–4601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hufeldt, M.R.; Nielsen, D.S.; Vogensen, F.K.; Midtvedt, T.; Hansen, A.K. Variation in the gut microbiota of laboratory mice is related to both genetic and environmental factors. Comp. Med. 2010, 60, 336–347. [Google Scholar] [PubMed]
- Macfarlane, G.T.; Macfarlane, S. Models for intestinal fermentation: Association between food components, delivery systems, bioavailability and functional interactions in the gut. Curr. Opin. Biotechnol. 2007, 18, 156–162. [Google Scholar] [CrossRef] [PubMed]
- Sekirov, I.; Russell, S.L.; Antunes, L.C.M.; Finlay, B.B.; Galla, S.; Chakraborty, S.; Cheng, X.; Yeo, J.; Mell, B.; Zhang, H.; et al. Gut Microbiota in Health and Disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.L.A.; Vieira-Silva, S.; Liston, A.; Raes, J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015, 8, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Chung, H.; Pamp, S.J.; Hill, J.A.; Surana, N.K.; Edelman, S.M.; Troy, E.B.; Reading, N.C.; Villablanca, E.J.; Wang, S.; Mora, J.R.; et al. Gut Immune Maturation Depends on Colonization with a Host-Specific Microbiota. Cell 2012, 149, 1578–1593. [Google Scholar] [CrossRef] [Green Version]
- Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The Effect of Diet on the Human Gut Microbiome: A Metagenomic Analysis in Humanized Gnotobiotic Mice. Sci. Transl. Med. 2009, 1, 6ra14. [Google Scholar] [CrossRef] [Green Version]
- Arrieta, M.-C.; Walter, J.; Finlay, B.B. Human Microbiota-Associated Mice: A Model with Challenges. Cell Host Microbe 2016, 19, 575–578. [Google Scholar] [CrossRef] [Green Version]
- Ericsson, A.C.; Franklin, C.L. Manipulating the Gut Microbiota: Methods and Challenges: Figure. ILAR J. 2015, 56, 205–217. [Google Scholar] [CrossRef] [Green Version]
- Denning, T.L.; Norris, B.A.; Medina-Contreras, O.; Manicassamy, S.; Geem, D.; Madan, R.; Karp, C.L.; Pulendran, B. Functional Specializations of Intestinal Dendritic Cell and Macrophage Subsets That Control Th17 and Regulatory T Cell Responses Are Dependent on the T Cell/APC Ratio, Source of Mouse Strain, and Regional Localization. J. Immunol. 2011, 187, 733–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ericsson, A.C.; Davis, J.W.; Spollen, W.; Bivens, N.; Givan, S.; Hagan, C.E.; McIntosh, M.; Franklin, C.L. Effects of Vendor and Genetic Background on the Composition of the Fecal Microbiota of Inbred Mice. PLoS ONE 2015, 10, e0116704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ivanov, I.I.; de Llanos Frutos, R.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific Microbiota Direct the Differentiation of IL-17-Producing T-Helper Cells in the Mucosa of the Small Intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macfarlane, G.; Macfarlane, S.; Gibson, G. Validation of a Three-Stage Compound Continuous Culture System for Investigating the Effect of Retention Time on the Ecology and Metabolism of Bacteria in the Human Colon. Microb. Ecol. 1998, 35, 180–187. [Google Scholar] [CrossRef] [PubMed]
- Van den Abbeele, P.; Grootaert, C.; Marzorati, M.; Possemiers, S.; Verstraete, W.; Gérard, P.; Rabot, S.; Bruneau, A.; El Aidy, S.; Derrien, M.; et al. Microbial Community Development in a Dynamic Gut Model Is Reproducible, Colon Region Specific, and Selective for Bacteroidetes and Clostridium Cluster IX. Appl. Environ. Microbiol. 2010, 76, 5237–5246. [Google Scholar] [CrossRef] [Green Version]
- Shultz, L.D.; Brehm, M.A.; Garcia-Martinez, J.V.; Greiner, D.L. Humanized mice for immune system investigation: Progress, promise and challenges. Nat. Rev. Immunol. 2012, 12, 786–798. [Google Scholar] [CrossRef]
- Weger, B.D.; Gobet, C.; Yeung, J.; Martin, E.; Jimenez, S.; Betrisey, B.; Foata, F.; Berger, B.; Balvay, A.; Foussier, A.; et al. The Mouse Microbiome Is Required for Sex-Specific Diurnal Rhythms of Gene Expression and Metabolism. Cell Metab. 2019, 29, 362–382.e8. [Google Scholar] [CrossRef] [Green Version]
- Gustafsson, B. Germ-free rearing of rats. Acta Anat. 1946, 2, 376–391. [Google Scholar] [CrossRef]
- Al-Asmakh, M.; Zadjali, F. Use of Germ-Free Animal Models in Microbiota-Related Research. J. Microbiol. Biotechnol. 2015, 25, 1583–1588. [Google Scholar] [CrossRef] [Green Version]
- Gordon, H.A.; Bruckner-Kardoss, E.; Wostmann, B.S. Aging in Germ-free Mice: Life Tables and Lesions Observed at Natural Death. J. Gerontol. 1966, 21, 380–387. [Google Scholar] [CrossRef]
- Bibiloni, R. Rodent models to study the relationships between mammals and their bacterial inhabitants. Gut Microbes 2012, 3, 536–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aluwihare, A.P.R. An ultrastructural study of the effect of neomycin on the colon in the human subject and in the conventional and the germ-free mouse. Gut 1971, 12, 341–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamamoto, Y.; Nakanishi, Y.; Murakami, S.; Aw, W.; Tsukimi, T.; Nozu, R.; Ueno, M.; Hioki, K.; Nakahigashi, K.; Hirayama, A.; et al. A Metabolomic-Based Evaluation of the Role of Commensal Microbiota throughout the Gastrointestinal Tract in Mice. Microorganisms 2018, 6, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawase, T.; Nagasawa, M.; Ikeda, H.; Yasuo, S.; Koga, Y.; Furuse, M. Gut microbiota of mice putatively modifies amino acid metabolism in the host brain. Br. J. Nutr. 2017, 117, 775–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, W.-H.; Sommer, F.; Falk-Paulsen, M.; Ulas, T.; Best, P.; Fazio, A.; Kachroo, P.; Luzius, A.; Jentzsch, M.; Rehman, A.; et al. Exposure to the gut microbiota drives distinct methylome and transcriptome changes in intestinal epithelial cells during postnatal development. Genome Med. 2018, 10, 27. [Google Scholar] [CrossRef]
- Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.-N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef]
- Arentsen, T.; Khalid, R.; Qian, Y.; Heijtz, R.D. Sex-dependent alterations in motor and anxiety-like behavior of aged bacterial peptidoglycan sensing molecule 2 knockout mice. Brain Behav. Immun. 2018, 67, 345–354. [Google Scholar] [CrossRef]
- Nishino, R.; Mikami, K.; Takahashi, H.; Tomonaga, S.; Furuse, M.; Hiramoto, T.; Aiba, Y.; Koga, Y.; Sudo, N. Commensal microbiota modulate murine behaviors in a strictly contamination-free environment confirmed by culture-based methods. Neurogastroenterol. Motil. 2013, 25, 521-e371. [Google Scholar] [CrossRef]
- Charbonneau, M.R.; O’Donnell, D.; Blanton, L.V.; Totten, S.M.; Davis, J.C.C.; Barratt, M.J.; Cheng, J.; Guruge, J.; Talcott, M.; Bain, J.R.; et al. Sialylated Milk Oligosaccharides Promote Microbiota-Dependent Growth in Models of Infant Undernutrition. Cell 2016, 164, 859–871. [Google Scholar] [CrossRef] [Green Version]
- Luczynski, P.; McVey Neufeld, K.-A.; Oriach, C.S.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int. J. Neuropsychopharmacol. 2016, 19, pyw020. [Google Scholar] [CrossRef]
- Gordon, H.A.; Pesti, L. The gnotobiotic animal as a tool in the study of host microbial relationships. Bacteriol. Rev. 1971, 35, 390–429. [Google Scholar] [CrossRef] [PubMed]
- Lyte, J.; Proctor, A.; Phillips, G.; Lyte, M.; Wannemuehler, M. Altered Schaedler flora mice: A defined microbiota animal model to study the microbiota-gut-brain axis. Behav. Brain Res. 2019, 356, 221–226. [Google Scholar] [CrossRef] [PubMed]
- Brand, M.W.; Wannemuehler, M.J.; Phillips, G.J.; Proctor, A.; Overstreet, A.-M.; Jergens, A.E.; Orcutt, R.P.; Fox, J.G. The Altered Schaedler Flora: Continued Applications of a Defined Murine Microbial Community. ILAR J. 2015, 56, 169–178. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Guilherme, M.; Nguyen, V.; Reinhardt, C.; Endres, K. Impact of Gut Microbiome Manipulation in 5xFAD Mice on Alzheimer’s Disease-Like Pathology. Microorganisms 2021, 9, 815. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.-J.; Lee, K.-E.; Kim, J.-K.; Kim, D.-H. Suppression of gut dysbiosis by Bifidobacterium longum alleviates cognitive decline in 5XFAD transgenic and aged mice. Sci. Rep. 2019, 9, 11814. [Google Scholar] [CrossRef] [Green Version]
- Zhao, W.; Wang, J.; Latta, M.; Wang, C.; Liu, Y.; Ma, W.; Zhou, Z.; Hu, S.; Chen, P.; Liu, Y. Rhizoma Gastrodiae Water Extract Modulates the Gut Microbiota and Pathological Changes of P-TauThr231 to Protect Against Cognitive Impairment in Mice. Front. Pharmacol. 2022, 13, 903659. [Google Scholar] [CrossRef]
- Fasina, O.B.; Wang, J.; Mo, J.; Osada, H.; Ohno, H.; Pan, W.; Xiang, L.; Qi, J. Gastrodin From Gastrodia elata Enhances Cognitive Function and Neuroprotection of AD Mice via the Regulation of Gut Microbiota Composition and Inhibition of Neuron Inflammation. Front. Pharmacol. 2022, 13, 814271. [Google Scholar] [CrossRef] [PubMed]
- Mosaferi, B.; Jand, Y.; Salari, A.-A. Gut microbiota depletion from early adolescence alters anxiety and depression-related behaviours in male mice with Alzheimer-like disease. Sci. Rep. 2021, 11, 22941. [Google Scholar] [CrossRef]
- Lee, M.; Lee, S.-H.; Kim, M.-S.; Ahn, K.-S.; Kim, M. Effect of Lactobacillus dominance modified by Korean Red Ginseng on the improvement of Alzheimer’s disease in mice. J. Ginseng Res. 2020, 46, 464–472. [Google Scholar] [CrossRef]
- Sun, J.; Xu, J.; Ling, Y.; Wang, F.; Gong, T.; Yang, C.; Ye, S.; Ye, K.; Wei, D.; Song, Z.; et al. Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Transl. Psychiatry 2019, 9, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kundu, P.; Stagaman, K.; Kasschau, K.; Holden, S.; Shulzhenko, N.; Sharpton, T.J.; Raber, J. Fecal Implants From AppNL–G–F and AppNL–G–F/E4 Donor Mice Sufficient to Induce Behavioral Phenotypes in Germ-Free Mice. Front. Behav. Neurosci. 2022, 16, 791128. [Google Scholar] [CrossRef] [PubMed]
- Payne, L.E.; Gagnon, D.J.; Riker, R.R.; Seder, D.B.; Glisic, E.K.; Morris, J.G.; Fraser, G.L. Cefepime-induced neurotoxicity: A systematic review. Crit. Care 2017, 21, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Yulug, B.; Hanoglu, L.; Ozansoy, M.; Isık, D.; Kilic, U.; Kilic, E.; Schabitz, W.R. Therapeutic role of rifampicin in Alzheimer’s disease. Psychiatry Clin. Neurosci. 2018, 72, 152–159. [Google Scholar] [CrossRef] [Green Version]
- Tsai, G.E.; Falk, W.E.; Gunther, J.; Coyle, J.T. Improved Cognition in Alzheimer’s Disease With Short-Term D-Cycloserine Treatment. Am. J. Psychiatry 1999, 156, 467–469. [Google Scholar] [CrossRef]
- Loeb, M.B.; Molloy, D.W.; Smieja, M.; Standish, T.; Goldsmith, C.H.; Mahony, J.; Smith, S.; Borrie, M.; Decoteau, E.; Davidson, W.; et al. A Randomized, Controlled Trial of Doxycycline and Rifampin for Patients with Alzheimer’s Disease. J. Am. Geriatr. Soc. 2004, 52, 381–387. [Google Scholar] [CrossRef]
- Zhang, R.; Miller, R.G.; Gascon, R.; Champion, S.; Katz, J.; Lancero, M.; Narvaez, A.; Honrada, R.; Ruvalcaba, D.; McGrath, M.S. Circulating endotoxin and systemic immune activation in sporadic amyotrophic lateral sclerosis (sALS). J. Neuroimmunol. 2009, 206, 121–124. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Cong, L.; Jaber, V.; Lukiw, W.J. Microbiome-Derived Lipopolysaccharide Enriched in the Perinuclear Region of Alzheimer’s Disease Brain. Front. Immunol. 2017, 8, 1064. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Cong, L.; Lukiw, W.J. Lipopolysaccharide (LPS) Accumulates in Neocortical Neurons of Alzheimer’s Disease (AD) Brain and Impairs Transcription in Human Neuronal-Glial Primary Co-cultures. Front. Aging Neurosci. 2017, 9, 407. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Jaber, V.; Lukiw, W.J. Secretory Products of the Human GI Tract Microbiome and Their Potential Impact on Alzheimer’s Disease (AD): Detection of Lipopolysaccharide (LPS) in AD Hippocampus. Front. Cell. Infect. Microbiol. 2017, 7, 318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dave, M.; Higgins, P.D.; Middha, S.; Rioux, K.P. The human gut microbiome: Current knowledge, challenges, and future directions. Transl. Res. 2012, 160, 246–257. [Google Scholar] [CrossRef] [PubMed]
- Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dethlefsen, L.; Relman, D.A. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4554–4561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The Pervasive Effects of an Antibiotic on the Human Gut Microbiota, as Revealed by Deep 16S rRNA Sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
- Spor, A.; Koren, O.; Ley, R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat. Rev. Microbiol. 2011, 9, 279–290. [Google Scholar] [CrossRef]
- Walker, A.; Ince, J.; Duncan, S.H.; Webster, L.M.; Holtrop, G.; Ze, X.; Brown, D.; Stares, M.D.; Scott, P.; Bergerat, A.; et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2010, 5, 220–230. [Google Scholar] [CrossRef] [PubMed]
- Macfarlane, G.; Gibson, G.; Cummings, J. Comparison of fermentation reactions in different regions of the human colon. J. Appl. Bacteriol. 1992, 72, 57–64. [Google Scholar] [CrossRef]
- Salzman, N.H.; Bevins, C.L. Negative Interactions with the Microbiota: IBD. Adv. Exp. Med. Biol. 2008, 635, 67–78. [Google Scholar] [CrossRef]
- Verhaar, B.J.H.; Hendriksen, H.M.A.; de Leeuw, F.A.; Doorduijn, A.S.; van Leeuwenstijn, M.; Teunissen, C.E.; Barkhof, F.; Scheltens, P.; Kraaij, R.; van Duijn, C.M.; et al. Gut Microbiota Composition Is Related to AD Pathology. Front. Immunol. 2021, 12, 794519. [Google Scholar] [CrossRef]
- Park, S.-H.; Lee, J.H.; Shin, J.; Kim, J.-S.; Cha, B.; Lee, S.; Kwon, K.S.; Shin, Y.W.; Choi, S.H. Cognitive function improvement after fecal microbiota transplantation in Alzheimer’s dementia patient: A case report. Curr. Med Res. Opin. 2021, 37, 1739–1744. [Google Scholar] [CrossRef] [PubMed]
- Hazan, S. Rapid improvement in Alzheimer’s disease symptoms following fecal microbiota transplantation: A case report. J. Int. Med Res. 2020, 48, 0300060520925930. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.-S.; Cha, L.; Sim, M.; Jung, S.; Chun, W.Y.; Baik, H.W.; Shin, D.-M. Probiotic Supplementation Improves Cognitive Function and Mood with Changes in Gut Microbiota in Community-Dwelling Older Adults: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. J. Gerontol. Ser. A 2021, 76, 32–40. [Google Scholar] [CrossRef] [PubMed]
- Leblhuber, F.; Steiner, K.; Schuetz, B.; Fuchs, D.; Gostner, J.M. Probiotic Supplementation in Patients with Alzheimer’s Dementia—An Explorative Intervention Study. Curr. Alzheimer Res. 2018, 15, 1106–1113. [Google Scholar] [CrossRef]
- Rajanala, K.; Kumar, N.; Chamallamudi, M.R. Modulation of Gut-Brain Axis by Probiotics: A Promising Anti-depressant Approach. Curr. Neuropharmacol. 2021, 19, 990–1006. [Google Scholar] [CrossRef]
- Xu, M.; Mo, X.; Huang, H.; Chen, X.; Liu, H.; Peng, Z.; Chen, L.; Rong, S.; Yang, W.; Xu, S.; et al. Yeast β-glucan alleviates cognitive deficit by regulating gut microbiota and metabolites in Aβ1–42-induced AD-like mice. Int. J. Biol. Macromol. 2020, 161, 258–270. [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]
- Xin, Y.; Diling, C.; Jian, Y.; Ting, L.; Guoyan, H.; Hualun, L.; Xiaocui, T.; Guoxiao, L.; Ou, S.; Chaoqun, Z.; et al. Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice. Front. Neurol. 2018, 9, 412. [Google Scholar] [CrossRef]
- Deng, S.; Chen, C.; Lin, H.; Cheng, I.H. The beneficial effect of synbiotics consumption on Alzheimer’s disease mouse model via reducing local and systemic inflammation. IUBMB Life 2021, 74, 748–753. [Google Scholar] [CrossRef]
- Gu, Y.; Nishikawa, M.; Brickman, A.M.; Manly, J.J.; Schupf, N.; Mayeux, R.P. Association of Dietary Prebiotic Consumption with Reduced Risk of Alzheimer’s Disease in a Multiethnic Population. Curr. Alzheimer Res. 2021, 18, 984–992. [Google Scholar] [CrossRef]
- Barbosa, R.S.D.; Vieira-Coelho, M.A. Probiotics and prebiotics: Focus on psychiatric disorders—A systematic review. Nutr. Rev. 2020, 78, 437–450. [Google Scholar] [CrossRef] [PubMed]
- Lilly, D.M.; Stillwell, R.H. Probiotics: Growth-Promoting Factors Produced by Microorganisms. Science 1965, 147, 747–748. [Google Scholar] [CrossRef] [PubMed]
- Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collado, M.C.; Gueimonde, M.; Sanz, Y.; Salminen, S. Adhesion Properties and Competitive Pathogen Exclusion Ability of Bifidobacteria with Acquired Acid Resistance. J. Food Prot. 2006, 69, 1675–1679. [Google Scholar] [CrossRef]
- Collado, M.C.; Hernández, M.; Sanz, Y. Production of Bacteriocin-Like Inhibitory Compounds by Human Fecal Bifidobacterium Strains. J. Food Prot. 2005, 68, 1034–1040. [Google Scholar] [CrossRef]
- Martinez, F.A.C.; Balciunas, E.M.; Converti, A.; Cotter, P.D.; de Souza Oliveira, R.P. Bacteriocin production by Bifidobacterium spp. A review. Biotechnol. Adv. 2013, 31, 482–488. [Google Scholar] [CrossRef]
- Bajaj, B.K.; Claes, I.J.; Lebeer, S. Functional mechanisms of probiotics. J. Microbiol. Biotechnol. Food Sci. 2021, 2021, 321–327. [Google Scholar]
- Sichetti, M.; De Marco, S.; Pagiotti, R.; Traina, G.; Pietrella, D. Anti-inflammatory effect of multistrain probiotic formulation (L. rhamnosus, B. lactis, and B. longum). Nutrition 2018, 53, 95–102. [Google Scholar] [CrossRef]
- Distrutti, E.; O’Reilly, J.-A.; McDonald, C.; Cipriani, S.; Renga, B.; Lynch, M.A.; Fiorucci, S. Modulation of Intestinal Microbiota by the Probiotic VSL#3 Resets Brain Gene Expression and Ameliorates the Age-Related Deficit in LTP. PLoS ONE 2014, 9, e106503. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- O’Hagan, C.; Li, J.V.; Marchesi, J.R.; Plummer, S.; Garaiova, I.; Good, M.A. Long-term multi-species Lactobacillus and Bifidobacterium dietary supplement enhances memory and changes regional brain metabolites in middle-aged rats. Neurobiol. Learn. Mem. 2017, 144, 36–47. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, Y.; Sugahara, H.; Shimada, K.; Mitsuyama, E.; Kuhara, T.; Yasuoka, A.; Kondo, T.; Abe, K.; Xiao, J.-Z. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer’s disease. Sci. Rep. 2017, 7, 13510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azm, S.A.N.; Djazayeri, A.; Safa, M.; Azami, K.; Ahmadvand, B.; Sabbaghziarani, F.; Sharifzadeh, M.; Vafa, M. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-amyloid (1–42) injected rats. Appl. Physiol. Nutr. Metab. 2018, 43, 718–726. [Google Scholar] [CrossRef]
- di Vito, R.; Conte, C.; Traina, G. A Multi-Strain Probiotic Formulation Improves Intestinal Barrier Function by the Modulation of Tight and Adherent Junction Proteins. Cells 2022, 11, 2617. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Klingbeil, E.A.; de La Serre, C.B. Microbiota modulation by eating patterns and diet composition: Impact on food intake. Am. J. Physiol. Integr. Comp. Physiol. 2018, 315, R1254–R1260. [Google Scholar] [CrossRef] [Green Version]
- Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; De Lorenzo, A. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2021, 13, 7. [Google Scholar] [CrossRef] [PubMed]
- Fung, T.T.; Rexrode, K.M.; Mantzoros, C.S.; Manson, J.E.; Willett, W.C.; Hu, F.B. Mediterranean Diet and Incidence of and Mortality From Coronary Heart Disease and Stroke in Women. Circulation 2009, 119, 1093–1100. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Garcia, E.; Rodriguez-Artalejo, F.; Li, T.Y.; Fung, T.T.; Li, S.; Willett, W.C.; Rimm, E.B.; Hu, F.B. The Mediterranean-style dietary pattern and mortality among men and women with cardiovascular disease. Am. J. Clin. Nutr. 2014, 99, 172–180. [Google Scholar] [CrossRef] [Green Version]
- Sofi, F.; Macchi, C.; Abbate, R.; Gensini, G.F.; Casini, A. Mediterranean diet and health. Biofactors 2013, 39, 335–342. [Google Scholar] [CrossRef]
- Martínez-Lapiscina, E.H.; Clavero, P.; Toledo, E.; Estruch, R.; Salas-Salvadó, J.; San Julián, B.; Sanchez-Tainta, A.; Ros, E.; Valls-Pedret, C.; Martinez-Gonzalez, M.Á. Mediterranean diet improves cognition: The PREDIMED-NAVARRA randomised trial. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1318–1325. [Google Scholar] [CrossRef] [Green Version]
- Valls-Pedret, C.; Sala-Vila, A.; Serra-Mir, M.; Corella, D.; De La Torre, R.; Martínez-González, M.Á.; Martínez-Lapiscina, E.H.; Fitó, M.; Pérez-Heras, A.; Salas-Salvadó, J.; et al. Mediterranean Diet and Age-Related Cognitive Decline: A Randomized Clinical Trial. JAMA Intern. Med. 2015, 175, 1094–1103. [Google Scholar] [CrossRef] [Green Version]
- Marseglia, A.; Xu, W.; Fratiglioni, L.; Fabbri, C.; Berendsen, A.A.M.; Bialecka-Debek, A.; Jennings, A.; Gillings, R.; Meunier, N.; Caumon, E.; et al. Effect of the NU-AGE Diet on Cognitive Functioning in Older Adults: A Randomized Controlled Trial. Front. Physiol. 2018, 9, 349. [Google Scholar] [CrossRef] [PubMed]
- Bartochowski, Z.; Conway, J.; Wallach, Y.; Chakkamparambil, B.; Alakkassery, S.; Grossberg, G.T. Dietary Interventions to Prevent or Delay Alzheimer’s Disease: What the Evidence Shows. Curr. Nutr. Rep. 2020, 9, 210–225. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.H.; Gao, X.; Na, M.; Kris-Etherton, P.; Mitchell, D.C.; Jensen, G.L. Dietary pattern, diet quality, and dementia: A systematic review and meta-analysis of prospective cohort studies. J. Alzheimers Dis. 2020, 78, 151–158. [Google Scholar] [CrossRef] [PubMed]
- Barbaresko, J.; Lellmann, A.W.; Schmidt, A.; Lehmann, A.; Amini, A.M.; Egert, S.; Schlesinger, S.; Nöthlings, U. Dietary Factors and Neurodegenerative Disorders: An Umbrella Review of Meta-Analyses of Prospective Studies. Adv. Nutr. Int. Rev. J. 2020, 11, 1161–1173. [Google Scholar] [CrossRef] [PubMed]
- Prinelli, F.; Fratiglioni, L.; Kalpouzos, G.; Musicco, M.; Adorni, F.; Johansson, I.; Marseglia, A.; Xu, W. Specific nutrient patterns are associated with higher structural brain integrity in dementia-free older adults. Neuroimage 2019, 199, 281–288. [Google Scholar] [CrossRef] [PubMed]
- Kao, Y.-C.; Ho, P.-C.; Tu, Y.-K.; Jou, I.-M.; Tsai, K.-J. Lipids and Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 1505. [Google Scholar] [CrossRef] [PubMed]
- Rainey-Smith, S.R.; Gu, Y.; Gardener, S.L.; Doecke, J.D.; Villemagne, V.L.; Brown, B.M.; Taddei, K.; Laws, S.M.; Sohrabi, H.R.; Weinborn, M.; et al. Mediterranean diet adherence and rate of cerebral Aβ-amyloid accumulation: Data from the Australian Imaging, Biomarkers and Lifestyle Study of Ageing. Transl. Psychiatry 2018, 8, 238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ułamek-Kozioł, M.; Czuczwar, S.J.; Januszewski, S.; Pluta, R. Ketogenic Diet and Epilepsy. Nutrients 2019, 11, 2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bostock, E.C.S.; Kirkby, K.C.; Taylor, B.V.M. The Current Status of the Ketogenic Diet in Psychiatry. Front. Psychiatry 2017, 8, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rusek, M.; Pluta, R.; Ułamek-Kozioł, M.; Czuczwar, S.J. Ketogenic Diet in Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 3892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Masino, S.A.; Rho, J.M. Mechanisms of ketogenic diet action. Epilepsia 2010, 51, 85. [Google Scholar] [CrossRef]
- Kashiwaya, Y.; Bergman, C.; Lee, J.-H.; Wan, R.; King, M.T.; Mughal, M.R.; Okun, E.; Clarke, K.; Mattson, M.P.; Veech, R.L. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2013, 34, 1530–1539. [Google Scholar] [CrossRef] [Green Version]
- Rojas-Morales, P.; Pedraza-Chaverri, J.; Tapia, E. Ketone bodies, stress response, and redox homeostasis. Redox Biol. 2020, 29, 101395. [Google Scholar] [CrossRef]
- Taylor, M.K.; Sullivan, D.K.; Mahnken, J.D.; Burns, J.; Swerdlow, R.H. Feasibility and efficacy data from a ketogenic diet intervention in Alzheimer’s disease. Alzheimer’s Dementia Transl. Res. Clin. Interv. 2018, 4, 28–36. [Google Scholar] [CrossRef]
- Włodarek, D. The possibility of use of the ketogenic diet and medium chain triglycerides supplementation in the support therapy of Alzheimer disease. Curr. Opin. Clin. Nutr. Metab. Care 2021, 24, 385–391. [Google Scholar] [CrossRef]
- Gupta, S.; Allen-Vercoe, E.; Petrof, E.O. Fecal microbiota transplantation: In perspective. Ther. Adv. Gastroenterol. 2016, 9, 229–239. [Google Scholar] [CrossRef] [Green Version]
- Surawicz, C.M.; Brandt, L.J.; Binion, D.G.; Ananthakrishnan, A.N.; Curry, S.R.; Gilligan, P.H.; McFarland, L.V.; Mellow, M.; Zuckerbraun, B.S. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile in-fections. Am. J. Gastroenterol. 2013, 108, 478–498. [Google Scholar] [CrossRef]
- Kim, N.; Jeon, S.H.; Ju, I.G.; Gee, M.S.; Do, J.; Oh, M.S.; Lee, J.K. Transplantation of gut microbiota derived from Alzheimer’s disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav. Immun. 2021, 98, 357–365. [Google Scholar] [CrossRef]
- Wang, M.; Cao, J.; Gong, C.; Amakye, W.K.; Yao, M.; Ren, J. Exploring the microbiota-Alzheimer’s disease linkage using short-term antibiotic treatment followed by fecal microbiota transplantation. Brain Behav. Immun. 2021, 96, 227–238. [Google Scholar] [CrossRef] [PubMed]
- Fujii, Y.; Nguyen, T.T.T.; Fujimura, Y.; Kameya, N.; Nakamura, S.; Arakawa, K.; Morita, H. Fecal metabolite of a gnotobiotic mouse transplanted with gut microbiota from a patient with Alzheimer’s disease. Biosci. Biotechnol. Biochem. 2019, 83, 2144–2152. [Google Scholar] [CrossRef] [PubMed]
- Dodiya, H.B.; Kuntz, T.; Shaik, S.M.; Baufeld, C.; Leibowitz, J.; Zhang, X.; Gottel, N.; Zhang, X.; Butovsky, O.; Gilbert, J.A.; et al. Sex-specific effects of microbiome perturbations on cerebral Aβ amyloidosis and microglia phenotypes. J. Exp. Med. 2019, 216, 1542–1560. [Google Scholar] [CrossRef] [Green Version]
- Gubert, C.; Choo, J.M.; Love, C.J.; Kodikara, S.; Masson, B.A.; Liew, J.J.M.; Wang, Y.; Kong, G.; Narayana, V.K.; Renoir, T.; et al. Faecal microbiota transplant ameliorates gut dysbiosis and cognitive deficits in Huntington’s disease mice. Brain Commun. 2022, 4, fcac205. [Google Scholar] [CrossRef]
- Choi, H.H.; Cho, Y.-S. Fecal Microbiota Transplantation: Current Applications, Effectiveness, and Future Perspectives. Clin. Endosc. 2016, 49, 257–265. [Google Scholar] [CrossRef] [PubMed]
- Schmulson, M.; Bashashati, M. Fecal microbiota transfer for bowel disorders: Efficacy or hype? Curr. Opin. Pharmacol. 2018, 43, 72–80. [Google Scholar] [CrossRef] [PubMed]
- Tan, P.; Li, X.; Shen, J.; Feng, Q. Fecal Microbiota Transplantation for the Treatment of Inflammatory Bowel Disease: An Update. Front. Pharmacol. 2020, 11, 574533. [Google Scholar] [CrossRef]
Advantages | Disadvantages |
---|---|
|
|
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. |
© 2023 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
Nguyen, N.M.; Cho, J.; Lee, C. Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship. Int. J. Mol. Sci. 2023, 24, 4047. https://doi.org/10.3390/ijms24044047
Nguyen NM, Cho J, Lee C. Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship. International Journal of Molecular Sciences. 2023; 24(4):4047. https://doi.org/10.3390/ijms24044047
Chicago/Turabian StyleNguyen, Ngoc Minh, Jungsook Cho, and Choongho Lee. 2023. "Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship" International Journal of Molecular Sciences 24, no. 4: 4047. https://doi.org/10.3390/ijms24044047
APA StyleNguyen, N. M., Cho, J., & Lee, C. (2023). Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship. International Journal of Molecular Sciences, 24(4), 4047. https://doi.org/10.3390/ijms24044047