Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases
Abstract
:1. Introduction
2. The Gut–Microbiota–Brain Axis in Health and Neurodegeneration
3. Lactic Acid Bacteria (LAB), Lactiplantibacillus plantarum, and Neurodegenerative Diseases
4. Alzheimer’s Disease, Cognitive Impairment, and Aging
5. Parkinson’s Disease (PD)
6. Multiple Sclerosis (MS)
7. Amyotrophic Lateral Sclerosis (ALS)
8. Autism Spectrum Disorder (ASD)
9. Anxiety and Depression
10. Conclusions, Limitations, and Future Perspective
Author Contributions
Funding
Conflicts of Interest
Abbreviations
Aβ | Amyloid-beta |
AD | Alzheimer disease |
ALS | Amyotrophic lateral sclerosis |
ASD | Autism spectrum disorder |
BBB | Blood–brain barrier |
BMI | Body mass index |
CNS | Central nervous system |
CXCL2 | C-X-C motif chemokine ligand 2 |
FMT | Fecal microbiota transplantation |
GABA | Gamma amino-butyric acid |
GF | Germ-free |
GI | Gastrointestinal |
GM | Gut microbiota |
IgA | Immunoglobulin A |
IL6 | Interleukin-6 |
LAB | Lactic acid bacteria |
LBP | Lactiplantibacillus plantarum |
LPS | Lipopolysaccharides |
MS | Multiple sclerosis |
NLRP-3 | Leucine rich repeat and pyrin domain containing-3 |
Nrf2 | Nuclear factor-E2-related factor 2 |
PD | Parkinson disease |
SCFAs | Short-chain fatty acids |
References
- Ding, C.; Wu, Y.; Chen, X.; Chen, Y.; Kang, D.; Fang, W.; Chen, F. Global, regional, and national burden and attributable risk factors of neurological disorders: The Global Burden of Disease study 1990–2019. Front. Public Health 2022, 10, 952161. [Google Scholar] [CrossRef] [PubMed]
- Carloni, S.; Bertocchi, A.; Mancinelli, S.; Bellini, M.; Erreni, M.; Borreca, A.; Braga, D.; Giugliano, S.; Mozzarelli, A.M.; Manganaro, D.; et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science 2021, 374, 439–448. [Google Scholar] [CrossRef] [PubMed]
- Zlokovic, B.V. The Blood-Brain Barrier in Health and Chronic Neurodegenerative Disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef]
- Boeckxstaens, G.; Camilleri, M.; Sifrim, D.; Houghton, L.A.; Elsenbruch, S.; Lindberg, G.; Azpiroz, F.; Parkman, H.P. Fundamentals of Neurogastroenterology: Physiology/Motility—Sensation. Gastroenterology 2016, 150, 1292–1304.e2. [Google Scholar] [CrossRef]
- Bengmark, S. Ecological control of the gastrointestinal tract. The role of probiotic flora. Gut 1998, 42, 2–7. [Google Scholar] [CrossRef] [PubMed]
- Niess, J.H.; Brand, S.; Gu, X.; Landsman, L.; Jung, S.; McCormick, B.A.; Vyas, J.M.; Boes, M.; Ploegh, H.L.; Fox, J.G.; et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 2005, 307, 254–258. [Google Scholar] [CrossRef]
- Mowat, A.M.; Maloy, K.J.; Donachie, A.M. Immune-stimulating complexes as adjuvants for inducing local and systemic immunity after oral immunization with protein antigens. Immunology 1993, 80, 527–534. [Google Scholar]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
- Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
- Arrieta, M.-C.; Stiemsma, L.T.; Amenyogbe, N.; Brown, E.M.; Finlay, B. The Intestinal Microbiome in Early Life: Health and Disease. Front. Immunol. 2014, 5, 427. [Google Scholar] [CrossRef]
- Laursen, M.F.; Bahl, M.I.; Michaelsen, K.F.; Licht, T.R. First Foods and Gut Microbes. Front. Microbiol. 2017, 8, 356. [Google Scholar] [CrossRef] [PubMed]
- Hollister, E.B.; Riehle, K.; Luna, R.A.; Weidler, E.M.; Rubio-Gonzales, M.; Mistretta, T.-A.; Raza, S.; Doddapaneni, H.V.; Metcalf, G.A.; Muzny, D.M.; et al. Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome 2015, 3, 36. [Google Scholar] [CrossRef] [PubMed]
- Bien, J.; Palagani, V.; Bozko, P. The intestinal microbiota dysbiosis and Clostridium difficile infection: Is there a relationship with inflammatory bowel disease? Ther. Adv. Gastroenterol. 2013, 6, 53–68. [Google Scholar] [CrossRef]
- Knights, D.; Lassen, K.G.; Xavier, R.J. Advances in inflammatory bowel disease pathogenesis: Linking host genetics and the microbiome. Gut 2013, 62, 1505–1510. [Google Scholar] [CrossRef] [PubMed]
- Arnold, J.W.; Roach, J.; Azcarate-Peril, M.A. Emerging Technologies for Gut Microbiome Research. Trends Microbiol. 2016, 24, 887–901. [Google Scholar] [CrossRef]
- Bull, M.J.; Plummer, N.T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. Clin. J. 2014, 13, 17–22. [Google Scholar]
- Rath, C.M.; Dorrestein, P.C. The bacterial chemical repertoire mediates metabolic exchange within gut microbiomes. Curr. Opin. Microbiol. 2012, 15, 147–154. [Google Scholar] [CrossRef]
- Hvas, C.L.; Dahl Jørgensen, S.M.; Jørgensen, S.P.; Storgaard, M.; Lemming, L.; Hansen, M.M.; Erikstrup, C.; Dahlerup, J.F. Fecal Microbiota Transplantation Is Superior to Fidaxomicin for Treatment of Recurrent Clostridium difficile Infection. Gastroenterology 2019, 156, 1324–1332.e3. [Google Scholar] [CrossRef]
- Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-Gut Microbiota Metabolic Interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef]
- Lange, K.; Buerger, M.; Stallmach, A.; Bruns, T. Effects of Antibiotics on Gut Microbiota. Dig. Dis. 2016, 34, 260–268. [Google Scholar] [CrossRef]
- Zhang, Y.-J.; Li, S.; Gan, R.-Y.; Zhou, T.; Xu, D.-P.; Li, H.-B. Impacts of Gut Bacteria on Human Health and Diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519. [Google Scholar] [CrossRef] [PubMed]
- Lyte, M. Microbial endocrinology: Host-microbiota neuroendocrine interactions influencing brain and behavior. Gut Microbes 2014, 5, 381–389. [Google Scholar] [CrossRef] [PubMed]
- Mu, G.; Gao, Y.; Tuo, Y.; Li, H.; Zhang, Y.; Qian, F.; Jiang, S. Assessing and comparing antioxidant activities of lactobacilli strains by using different chemical and cellular antioxidant methods. J. Dairy Sci. 2018, 101, 10792–10806. [Google Scholar] [CrossRef] [PubMed]
- Xiao, M.; Xiao, Z.J.; Yang, B.; Lan, Z.; Fang, F. Blood-Brain Barrier: More Contributor to Disruption of Central Nervous System Homeostasis Than Victim in Neurological Disorders. Front. Neurosci. 2020, 14, 764. [Google Scholar] [CrossRef]
- 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]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef]
- Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
- Barrett, E.; Ross, R.P.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
- Kali, A. Psychobiotics: An emerging probiotic in psychiatric practice. Biomed. J. 2016, 39, 223–224. [Google Scholar] [CrossRef]
- Li, H.; Qiu, T.; Huang, G.; Cao, Y. Production of gamma-aminobutyric acid by Lactobacillus brevis NCL912 using fed-batch fermentation. Microb. Cell Factories 2010, 9, 85. [Google Scholar] [CrossRef]
- Mombereau, C.; Kaupmann, K.; Froestl, W.; Sansig, G.; van der Putten, H.; Cryan, J.F. Genetic and pharmacological evidence of a role for GABA(B) receptors in the modulation of anxiety- and antidepressant-like behavior. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2004, 29, 1050–1062. [Google Scholar] [CrossRef]
- Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Duan, Y.; Jin, F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef]
- Cheng, L.-H.; Liu, Y.-W.; Wu, C.-C.; Wang, S.; Tsai, Y.-C. Psychobiotics in mental health, neurodegenerative and neurodevelopmental disorders. J. Food Drug Anal. 2019, 27, 632–648. [Google Scholar] [CrossRef]
- Liu, W.-H.; Chuang, H.-L.; Huang, Y.-T.; Wu, C.-C.; Chou, G.-T.; Wang, S.; Tsai, Y.-C. Alteration of behavior and monoamine levels attributable to Lactobacillus plantarum PS128 in germ-free mice. Behav. Brain Res. 2016, 298, 202–209. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.-W.; Liu, W.-H.; Wu, C.-C.; Juan, Y.-C.; Wu, Y.-C.; Tsai, H.-P.; Wang, S.; Tsai, Y.-C. Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naïve adult mice. Brain Res. 2016, 1631, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef]
- Sherman, M.P.; Bennett, S.H.; Hwang, F.F.Y.; Yu, C. Neonatal small bowel epithelia: Enhancing anti-bacterial defense with lactoferrin and Lactobacillus GG. Biometals Int. J. Role Met. Ions Biol. Biochem. Med. 2004, 17, 285–289. [Google Scholar] [CrossRef]
- Bhattacharjee, S.; Lukiw, W.J. Alzheimer’s disease and the microbiome. Front. Cell. Neurosci. 2013, 7, 153. [Google Scholar] [CrossRef]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
- Brydges, C.R.; Fiehn, O.; Mayberg, H.S.; Schreiber, H.; Dehkordi, S.M.; Bhattacharyya, S.; Cha, J.; Choi, K.S.; Craighead, W.E.; Krishnan, R.R.; et al. Indoxyl sulfate, a gut microbiome-derived uremic toxin, is associated with psychic anxiety and its functional magnetic resonance imaging-based neurologic signature. Sci. Rep. 2021, 11, 21011. [Google Scholar] [CrossRef]
- Salminen, A. Activation of aryl hydrocarbon receptor (AhR) in Alzheimer’s disease: Role of tryptophan metabolites generated by gut host-microbiota. J. Mol. Med. 2023, 101, 201–222. [Google Scholar] [CrossRef]
- Bobot, M.; Thomas, L.; Moyon, A.; Fernandez, S.; McKay, N.; Balasse, L.; Garrigue, P.; Brige, P.; Chopinet, S.; Poitevin, S.; et al. Uremic Toxic Blood-Brain Barrier Disruption Mediated by AhR Activation Leads to Cognitive Impairment during Experimental Renal Dysfunction. J. Am. Soc. Nephrol. JASN 2020, 31, 1509–1521. [Google Scholar] [CrossRef]
- Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef]
- Valvassori, S.S.; Resende, W.R.; Budni, J.; Dal-Pont, G.C.; Bavaresco, D.V.; Réus, G.Z.; Carvalho, A.F.; Gonçalves, C.L.; Furlanetto, C.B.; Streck, E.L.; et al. Sodium Butyrate, a Histone Deacetylase Inhibitor, Reverses Behavioral and Mitochondrial Alterations in Animal Models of Depression Induced by Early- or Late-life Stress. Curr. Neurovasc. Res. 2015, 12, 312–320. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Melas, P.A.; Wegener, G.; Mathé, A.A.; Lavebratt, C. Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the Bdnf gene. Int. J. Neuropsychopharmacol. 2014, 18, pyu032. [Google Scholar] [CrossRef] [PubMed]
- Kabouridis, P.S.; Lasrado, R.; McCallum, S.; Chng, S.H.; Snippert, H.J.; Clevers, H.; Pettersson, S.; Pachnis, V. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron 2015, 85, 289–295. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
- Calvani, R.; Picca, A.; Lo Monaco, M.R.; Landi, F.; Bernabei, R.; Marzetti, E. Of Microbes and Minds: A Narrative Review on the Second Brain Aging. Front. Med. 2018, 5, 53. [Google Scholar] [CrossRef]
- Singh, N.; Singh, V.; Rai, S.N.; Mishra, V.; Vamanu, E.; Singh, M.P. Deciphering the gut microbiome in neurodegenerative diseases and metagenomic approaches for characterization of gut microbes. Biomed. Pharmacother. 2022, 156, 113958. [Google Scholar] [CrossRef]
- Askarova, S.; Umbayev, B.; Masoud, A.-R.; Kaiyrlykyzy, A.; Safarova, Y.; Tsoy, A.; Olzhayev, F.; Kushugulova, A. The Links Between the Gut Microbiome, Aging, Modern Lifestyle and Alzheimer’s Disease. Front. Cell. Infect. Microbiol. 2020, 10, 104. [Google Scholar] [CrossRef]
- Molinero, N.; Antón-Fernández, A.; Hernández, F.; Ávila, J.; Bartolomé, B.; Moreno-Arribas, M.V. Gut Microbiota, an Additional Hallmark of Human Aging and Neurodegeneration. Neuroscience 2023, 518, 141–161. [Google Scholar] [CrossRef]
- Fröhlich, E.E.; Farzi, A.; Mayerhofer, R.; Reichmann, F.; Jačan, A.; Wagner, B.; Zinser, E.; Bordag, N.; Magnes, C.; Fröhlich, E.; et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain. Behav. Immun. 2016, 56, 140–155. [Google Scholar] [CrossRef] [PubMed]
- Kim, N.; Yun, M.; Oh, Y.J.; Choi, H.-J. Mind-altering with the gut: Modulation of the gut-brain axis with probiotics. J. Microbiol. 2018, 56, 172–182. [Google Scholar] [CrossRef] [PubMed]
- Mörkl, S.; Butler, M.I.; Holl, A.; Cryan, J.F.; Dinan, T.G. Probiotics and the Microbiota-Gut-Brain Axis: Focus on Psychiatry. Curr. Nutr. Rep. 2020, 9, 171–182. [Google Scholar] [CrossRef]
- Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef] [PubMed]
- Gupta, N.; Yadav, V.K.; Gacem, A.; Al-Dossari, M.; Yadav, K.K.; Abd El-Gawaad, N.S.; Ben Khedher, N.; Choudhary, N.; Kumar, P.; Cavalu, S. Deleterious Effect of Air Pollution on Human Microbial Community and Bacterial Flora: A Short Review. Int. J. Environ. Res. Public Health 2022, 19, 15494. [Google Scholar] [CrossRef]
- Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef]
- Liao, J.-F.; Cheng, Y.-F.; You, S.-T.; Kuo, W.-C.; Huang, C.-W.; Chiou, J.-J.; Hsu, C.-C.; Hsieh-Li, H.-M.; Wang, S.; Tsai, Y.-C. Lactobacillus plantarum PS128 alleviates neurodegenerative progression in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse models of Parkinson’s disease. Brain Behav. Immun. 2020, 90, 26–46. [Google Scholar] [CrossRef]
- Food and Agriculture Organization of the United Nations (FAO). Publication Preview Page. FAO Documents. Available online: https://www.fao.org/documents/card/en?details=7c102d95-2fd5-5b22-8faf-f0b2e68dfbb6/ (accessed on 15 February 2024).
- Bosco, N.; Noti, M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021, 22, 289–303. [Google Scholar] [CrossRef]
- Nogal, A.; Valdes, A.M.; Menni, C. The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health. Gut Microbes 2021, 13, 1897212. [Google Scholar] [CrossRef]
- Rahman, M.M.; Islam, M.R.; Shohag, S.; Ahasan, M.T.; Sarkar, N.; Khan, H.; Hasan, A.M.; Cavalu, S.; Rauf, A. Microbiome in cancer: Role in carcinogenesis and impact in therapeutic strategies. Biomed. Pharmacother. 2022, 149, 112898. [Google Scholar] [CrossRef]
- Foglietta, F.; Serpe, L.; Canaparo, R. The Effective Combination between 3D Cancer Models and Stimuli-Responsive Nanoscale Drug Delivery Systems. Cells 2021, 10, 3295. [Google Scholar] [CrossRef]
- Jeong, S.; Kim, Y.; Park, S.; Lee, D.; Lee, J.; Hlaing, S.P.; Yoo, J.-W.; Rhee, S.H.; Im, E. Lactobacillus plantarum Metabolites Elicit Anticancer Effects by Inhibiting Autophagy-Related Responses. Molecules 2023, 28, 1890. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Xiao, H.; Su, Y.; Cheng, D.; Jia, Y.; Li, Y.; Yin, Q.; Gao, J.; Tang, Y.; Bai, Q. Synergistic Inhibitory Effect of Honey and Lactobacillus plantarum on Pathogenic Bacteria and Their Promotion of Healing in Infected Wounds. Pathogens 2023, 12, 501. [Google Scholar] [CrossRef] [PubMed]
- OjiNjideka Hemphill, N.; Pezley, L.; Steffen, A.; Elam, G.; Kominiarek, M.A.; Odoms-Young, A.; Kessee, N.; Hamm, A.; Tussing-Humphreys, L.; Koenig, M.D. Feasibility Study of Lactobacillus plantarum 299v Probiotic Supplementation in an Urban Academic Facility among Diverse Pregnant Individuals. Nutrients 2023, 15, 875. [Google Scholar] [CrossRef]
- Li, D.; Ke, Y.; Zhan, R.; Liu, C.; Zhao, M.; Zeng, A.; Shi, X.; Ji, L.; Cheng, S.; Pan, B.; et al. Trimethylamine-N-oxide promotes brain aging and cognitive impairment in mice. Aging Cell 2018, 17, e12768. [Google Scholar] [CrossRef]
- Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98–104. [Google Scholar] [CrossRef] [PubMed]
- Seddik, H.A.; Bendali, F.; Gancel, F.; Fliss, I.; Spano, G.; Drider, D. Lactobacillus plantarum and Its Probiotic and Food Potentialities. Probiotics Antimicrob. Proteins 2017, 9, 111–122. [Google Scholar] [CrossRef]
- Cammarota, G.; Ianiro, G.; Bibbò, S.; Gasbarrini, A. Gut microbiota modulation: Probiotics, antibiotics or fecal microbiota transplantation? Intern. Emerg. Med. 2014, 9, 365–373. [Google Scholar] [CrossRef]
- Ho, S.-T.; Hsieh, Y.-T.; Wang, S.-Y.; Chen, M.-J. Improving effect of a probiotic mixture on memory and learning abilities in d-galactose–treated aging mice. J. Dairy Sci. 2019, 102, 1901–1909. [Google Scholar] [CrossRef]
- Lin, S.-W.; Tsai, Y.-S.; Chen, Y.-L.; Wang, M.-F.; Chen, C.-C.; Lin, W.-H.; Fang, T.J. Lactobacillus plantarum GKM3 Promotes Longevity, Memory Retention, and Reduces Brain Oxidation Stress in SAMP8 Mice. Nutrients 2021, 13, 2860. [Google Scholar] [CrossRef]
- Qian, Y.; Zhang, J.; Zhou, X.; Yi, R.; Mu, J.; Long, X.; Pan, Y.; Zhao, X.; Liu, W. Lactobacillus plantarum CQPC11 Isolated from Sichuan Pickled Cabbages Antagonizes d-galactose-Induced Oxidation and Aging in Mice. Molecules 2018, 23, 3026. [Google Scholar] [CrossRef]
- Samtiya, M.; Puniya, A.K.; Puniya, M.; Shah, N.P.; Dhewa, T.; Vemuri, R. Probiotic Regulation to Modulate Aging Gut and Brain Health: A Concise Review. Bacteria 2022, 1, 250–265. [Google Scholar] [CrossRef]
- Wang, F.; Zhou, H.; Deng, L.; Wang, L.; Chen, J.; Zhou, X. Serine Deficiency Exacerbates Inflammation and Oxidative Stress via Microbiota-Gut-Brain Axis in D-Galactose-Induced Aging Mice. Mediat. Inflamm. 2020, 2020, e5821428. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Yu, D.; Xue, L.; Li, H.; Du, J. Probiotics modulate the microbiota–gut–brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm. Sin. B 2020, 10, 475–487. [Google Scholar] [CrossRef] [PubMed]
- Zaydi, A.I.; Lew, L.-C.; Hor, Y.-Y.; Jaafar, M.H.; Chuah, L.-O.; Yap, K.-P.; Azlan, A.; Azzam, G.; Liong, M.-T. Lactobacillus plantarum DR7 improved brain health in aging rats via the serotonin, inflammatory and apoptosis pathways. Benef. Microbes 2020, 11, 753–766. [Google Scholar] [CrossRef]
- Zhang, J.; Zhao, X.; Jiang, Y.; Zhao, W.; Guo, T.; Cao, Y.; Teng, J.; Hao, X.; Zhao, J.; Yang, Z. Antioxidant status and gut microbiota change in an aging mouse model as influenced by exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibetan kefir. J. Dairy Sci. 2017, 100, 6025–6041. [Google Scholar] [CrossRef]
- Zhou, X.; Sun, H.; Tan, F.; Yi, R.; Zhou, C.; Deng, Y.; Mu, J.; Zhao, X. Anti-aging effect of Lactobacillus plantarum HFY09-fermented soymilk on D-galactose-induced oxidative aging in mice through modulation of the Nrf2 signaling pathway. J. Funct. Foods 2021, 78, 104386. [Google Scholar] [CrossRef]
- Moqri, M.; Herzog, C.; Poganik, J.R.; Justice, J.; Belsky, D.W.; Higgins-Chen, A.; Moskalev, A.; Fuellen, G.; Cohen, A.A.; Bautmans, I.; et al. Biomarkers of aging for the identification and evaluation of longevity interventions. Cell 2023, 186, 3758–3775. [Google Scholar] [CrossRef]
- Radzievskiĭ, S.A.; Kapel’ko, V.I. The strength and rate of contraction of the myocardium of the cardiac ventricles. Biull. Eksp. Biol. Med. 1969, 67, 3–5. [Google Scholar]
- Abedin, M.M.; Chourasia, R.; Phukon, L.C.; Sarkar, P.; Ray, R.C.; Singh, S.P.; Rai, A.K. Lactic acid bacteria in the functional food industry: Biotechnological properties and potential applications. Crit. Rev. Food Sci. Nutr. 2023, 5, 1–19. [Google Scholar] [CrossRef]
- Axelsson, L. Lactic Acid Bacteria: Classification and Physiology. In Lactic Acid Bacteria; Salminen, S., Von Wright, A., Ouwehand, A., Eds.; CRC Press: Boca Raton, FL, USA, 2004; ISBN 978-0-8247-5332-0. [Google Scholar]
- Guarner, F.; Malagelada, J.-R. Gut flora in health and disease. Lancet Lond. Engl. 2003, 361, 512–519. [Google Scholar] [CrossRef] [PubMed]
- Alves, J.L.; Lemos, L.; Rodrigues, N.M.; Pereira, V.B.; Barros, P.A.V.; Canesso, M.C.C.; Guimarães, M.A.F.; Cara, D.C.; Miyoshi, A.; Azevedo, V.A.; et al. Immunomodulatory effects of different strains of Lactococcus lactis in DSS-induced colitis. Braz. J. Microbiol. 2023, 54, 1203–1215. [Google Scholar] [CrossRef] [PubMed]
- Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of Commensal Microflora by Toll-Like Receptors Is Required for Intestinal Homeostasis. Cell 2004, 118, 229–241. [Google Scholar] [CrossRef]
- Amaretti, A.; di Nunzio, M.; Pompei, A.; Raimondi, S.; Rossi, M.; Bordoni, A. Antioxidant properties of potentially probiotic bacteria: In vitro and in vivo activities. Appl. Microbiol. Biotechnol. 2013, 97, 809–817. [Google Scholar] [CrossRef] [PubMed]
- Ianniello, R.G.; Ricciardi, A.; Parente, E.; Tramutola, A.; Reale, A.; Zotta, T. Aeration and supplementation with heme and menaquinone affect survival to stresses and antioxidant capability of Lactobacillus casei strains. LWT—Food Sci. Technol. 2015, 60, 817–824. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef]
- Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
- Martino, M.E.; Bayjanov, J.R.; Caffrey, B.E.; Wels, M.; Joncour, P.; Hughes, S.; Gillet, B.; Kleerebezem, M.; van Hijum, S.A.F.T.; Leulier, F. Nomadic lifestyle of Lactobacillus plantarum revealed by comparative genomics of 54 strains isolated from different habitats. Environ. Microbiol. 2016, 18, 4974–4989. [Google Scholar] [CrossRef]
- Fidanza, M.; Panigrahi, P.; Kollmann, T.R. Lactiplantibacillus plantarum-Nomad and Ideal Probiotic. Front. Microbiol. 2021, 12, 712236. [Google Scholar] [CrossRef]
- Nordström, E.A.; Teixeira, C.; Montelius, C.; Jeppsson, B.; Larsson, N. Lactiplantibacillus plantarum 299v (LP299V®): Three decades of research. Benef. Microbes 2021, 12, 441–465. [Google Scholar] [CrossRef] [PubMed]
- Hütt, P.; Songisepp, E.; Rätsep, M.; Mahlapuu, R.; Kilk, K.; Mikelsaar, M. Impact of probiotic Lactobacillus plantarum TENSIA in different dairy products on anthropometric and blood biochemical indices of healthy adults. Benef. Microbes 2015, 6, 233–243. [Google Scholar] [CrossRef] [PubMed]
- Kujawa-Szewieczek, A.; Adamczak, M.; Kwiecień, K.; Dudzicz, S.; Gazda, M.; Więcek, A. The Effect of Lactobacillus plantarum 299v on the Incidence of Clostridium difficile Infection in High Risk Patients Treated with Antibiotics. Nutrients 2015, 7, 10179–10188. [Google Scholar] [CrossRef] [PubMed]
- McNicholl, A.G.; Molina-Infante, J.; Lucendo, A.J.; Calleja, J.L.; Pérez-Aisa, Á.; Modolell, I.; Aldeguer, X.; Calafat, M.; Comino, L.; Ramas, M.; et al. Probiotic supplementation with Lactobacillus plantarum and Pediococcus acidilactici for Helicobacter pylori therapy: A randomized, double-blind, placebo-controlled trial. Helicobacter 2018, 23, e12529. [Google Scholar] [CrossRef]
- Vicariotto, F.; Mogna, L.; Del Piano, M. Effectiveness of the two microorganisms Lactobacillus fermentum LF15 and Lactobacillus plantarum LP01, formulated in slow-release vaginal tablets, in women affected by bacterial vaginosis: A pilot study. J. Clin. Gastroenterol. 2014, 48 (Suppl. S1), S106–S112. [Google Scholar] [CrossRef]
- Barraza-Ortiz, D.A.; Pérez-López, N.; Medina-López, V.M.; Minero-Alfaro, J.I.; Zamarripa-Dorsey, F.; Fernández-Martínez, N.d.C.; Llorente-Ramón, A.; Ramos-Aguilar, G.A. Combination of a Probiotic and an Antispasmodic Increases Quality of Life and Reduces Symptoms in Patients with Irritable Bowel Syndrome: A Pilot Study. Dig. Dis. 2020, 39, 294–300. [Google Scholar] [CrossRef]
- Fedorak, R.N.; Feagan, B.G.; Hotte, N.; Leddin, D.; Dieleman, L.A.; Petrunia, D.M.; Enns, R.; Bitton, A.; Chiba, N.; Paré, P.; et al. The Probiotic VSL#3 Has Anti-inflammatory Effects and Could Reduce Endoscopic Recurrence After Surgery for Crohn’s Disease. Clin. Gastroenterol. Hepatol. 2015, 13, 928–935.e2. [Google Scholar] [CrossRef]
- Kaźmierczak-Siedlecka, K.; Folwarski, M.; Skonieczna-Żydecka, K.; Ruszkowski, J.; Makarewicz, W. The use of Lactobacillus plantarum 299v (DSM 9843) in cancer patients receiving home enteral nutrition-study protocol for a randomized, double-blind, and placebo-controlled trial. Nutr. J. 2020, 19, 98. [Google Scholar] [CrossRef]
- Alisi, A.; Bedogni, G.; Baviera, G.; Giorgio, V.; Porro, E.; Paris, C.; Giammaria, P.; Reali, L.; Anania, F.; Nobili, V. Randomised clinical trial: The beneficial effects of VSL#3 in obese children with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2014, 39, 1276–1285. [Google Scholar] [CrossRef]
- Bosch, M.; Fuentes, M.C.; Audivert, S.; Bonachera, M.A.; Peiró, S.; Cuñé, J. Lactobacillus plantarum CECT 7527, 7528 and 7529: Probiotic candidates to reduce cholesterol levels. J. Sci. Food Agric. 2014, 94, 803–809. [Google Scholar] [CrossRef]
- Miraghajani, M.; Dehsoukhteh, S.S.; Rafie, N.; Hamedani, S.G.; Sabihi, S.; Ghiasvand, R. Potential mechanisms linking probiotics to diabetes: A narrative review of the literature. Sao Paulo Med. J. 2017, 135, 169–178. [Google Scholar] [CrossRef]
- Ferreira, S.T.; Lourenco, M.V.; Oliveira, M.M.; De Felice, F.G. Soluble amyloid-b oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front. Cell. Neurosci. 2015, 9, 191. [Google Scholar] [CrossRef] [PubMed]
- Rudzki, L.; Ostrowska, L.; Pawlak, D.; Małus, A.; Pawlak, K.; Waszkiewicz, N.; Szulc, A. Probiotic Lactobacillus Plantarum 299v decreases kynurenine concentration and improves cognitive functions in patients with major depression: A double-blind, randomized, placebo controlled study. Psychoneuroendocrinology 2019, 100, 213–222. [Google Scholar] [CrossRef]
- Huang, H.-J.; Chen, J.-L.; Liao, J.-F.; Chen, Y.-H.; Chieu, M.-W.; Ke, Y.-Y.; Hsu, C.-C.; Tsai, Y.-C.; Hsieh-Li, H.M. Lactobacillus plantarum PS128 prevents cognitive dysfunction in Alzheimer’s disease mice by modulating propionic acid levels, glycogen synthase kinase 3 beta activity, and gliosis. BMC Complement. Med. Ther. 2021, 21, 259. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Tankou, S.K.; Regev, K.; Healy, B.C.; Tjon, E.; Laghi, L.; Cox, L.M.; Kivisäkk, P.; Pierre, I.V.; Hrishikesh, L.; Gandhi, R.; et al. A probiotic modulates the microbiome and immunity in multiple sclerosis. Ann. Neurol. 2018, 83, 1147–1161. [Google Scholar] [CrossRef]
- Xin, Z.; Xin, C.; Huo, J.; Liu, Q.; Dong, H.; Li, R.; Liu, Y. Neuroprotective Effect of a Multistrain Probiotic Mixture in SOD1G93A Mice by Reducing SOD1 Aggregation and Targeting the Microbiota-Gut-Brain Axis. Mol. Neurobiol. 2024. [Google Scholar] [CrossRef]
- Guo, M.; Li, R.; Wang, Y.; Ma, S.; Zhang, Y.; Li, S.; Zhang, H.; Liu, Z.; You, C.; Zheng, H. Lactobacillus plantarum ST-III modulates abnormal behavior and gut microbiota in a mouse model of autism spectrum disorder. Physiol. Behav. 2022, 257, 113965. [Google Scholar] [CrossRef]
- Chen, H.-M.; Kuo, P.-H.; Hsu, C.-Y.; Chiu, Y.-H.; Liu, Y.-W.; Lu, M.-L.; Chen, C.-H. Psychophysiological Effects of Lactobacillus plantarum PS128 in Patients with Major Depressive Disorder: A Preliminary 8-Week Open Trial. Nutrients 2021, 13, 3731. [Google Scholar] [CrossRef]
- Sultana, O.F.; Hia, R.A.; Reddy, P.H. A Combinational Therapy for Preventing and Delaying the Onset of Alzheimer’s Disease: A Focus on Probiotic and Vitamin Co-Supplementation. Antioxidants 2024, 13, 202. [Google Scholar] [CrossRef]
- Zhao, Y.; Yang, G.; Zhao, Z.; Wang, C.; Duan, C.; Gao, L.; Li, S. Antidepressant-like effects of Lactobacillus plantarum DP189 in a corticosterone-induced rat model of chronic stress. Behav. Brain Res. 2020, 395, 112853. [Google Scholar] [CrossRef]
- Mazzini, L.; Mogna, L.; De Marchi, F.; Amoruso, A.; Pane, M.; Aloisio, I.; Cionci, N.B.; Gaggìa, F.; Lucenti, A.; Bersano, E.; et al. Potential Role of Gut Microbiota in ALS Pathogenesis and Possible Novel Therapeutic Strategies. J. Clin. Gastroenterol. 2018, 52 (Suppl. S1), S68–S70. [Google Scholar] [CrossRef]
- Liu, Y.-W.; Liong, M.T.; Chung, Y.-C.E.; Huang, H.-Y.; Peng, W.-S.; Cheng, Y.-F.; Lin, Y.-S.; Wu, Y.-Y.; Tsai, Y.-C. Effects of Lactobacillus plantarum PS128 on Children with Autism Spectrum Disorder in Taiwan: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2019, 11, 820. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Yeo, S.; Kim, B.; Holzapfel, W.H.; Kang, H.-J. Lactiplantibacillus plantarum APsulloc 331261 (GTB1TM) attenuates depressive-like behavior and physiology via association with gut microbiota in BALB/c mice. J. Funct. Foods 2023, 107, 105692. [Google Scholar] [CrossRef]
- Chung, Y.-C.; Jin, H.-M.; Cui, Y.; Kim, D.S.; Jung, J.M.; Park, J.-I.; Jung, E.-S.; Choi, E.-K.; Chae, S.-W. Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J. Funct. Foods 2014, 10, 465–474. [Google Scholar] [CrossRef]
- Ghyselinck, J.; Verstrepen, L.; Moens, F.; Van Den Abbeele, P.; Bruggeman, A.; Said, J.; Smith, B.; Barker, L.A.; Jordan, C.; Leta, V.; et al. Influence of probiotic bacteria on gut microbiota composition and gut wall function in an in-vitro model in patients with Parkinson’s disease. Int. J. Pharm. X 2021, 3, 100087. [Google Scholar] [CrossRef] [PubMed]
- Parracho, H.M.R.T.; Gibson, G.R.; Knott, F.; Bosscher, D.; Kleerebezem, M.; McCartney, A.L. A double-blind, placebo-controlled, crossover-designed probiotic feeding study in children diagnosed with autistic spectrum disorders. Int. J. Probiotics Prebiotics 2010, 5, 69–74. [Google Scholar]
- Mallikarjuna, N.; Praveen, K.; Yellamma, K. Role of Lactobacillus plantarum MTCC1325 in membrane-bound transport ATPases system in Alzheimer’s disease-induced rat brain. BioImpacts BI 2016, 6, 203–209. [Google Scholar] [CrossRef]
- Salehipour, Z.; Haghmorad, D.; Sankian, M.; Rastin, M.; Nosratabadi, R.; Soltan Dallal, M.M.; Tabasi, N.; Khazaee, M.; Nasiraii, L.R.; Mahmoudi, M. Bifidobacterium animalis in combination with human origin of Lactobacillus plantarum ameliorate neuroinflammation in experimental model of multiple sclerosis by altering CD4+ T cell subset balance. Biomed. Pharmacother. 2017, 95, 1535–1548. [Google Scholar] [CrossRef]
- Miao, Z.; Chen, L.; Zhang, Y.; Zhang, J.; Zhang, H. Bifidobacterium animalis subsp. lactis Probio-M8 alleviates abnormal behavior and regulates gut microbiota in a mouse model suffering from autism. mSystems 2024, 9, e0101323. [Google Scholar] [CrossRef]
- Zhu, R.; Fang, Y.; Li, H.; Liu, Y.; Wei, J.; Zhang, S.; Wang, L.; Fan, R.; Wang, L.; Li, S.; et al. Psychobiotic Lactobacillus plantarum JYLP-326 relieves anxiety, depression, and insomnia symptoms in test anxious college via modulating the gut microbiota and its metabolism. Front. Immunol. 2023, 14, 1158137. [Google Scholar] [CrossRef] [PubMed]
- Bonfili, L.; Cecarini, V.; Gogoi, O.; Berardi, S.; Scarpona, S.; Angeletti, M.; Rossi, G.; Eleuteri, A.M. Gut microbiota manipulation through probiotics oral administration restores glucose homeostasis in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2020, 87, 35–43. [Google Scholar] [CrossRef] [PubMed]
- Sajedi, D.; Shabani, R.; Elmieh, A. Effects of exercise training with Lactobacillus plantarum intake on ionized calcium-binding adaptor molecule 1, glial fibrillary acidic protein, interleukin-6 and interleukin-1β in cuprizone-induced demyelination mouse model of multiple sclerosis. Clin. Exp. Neuroimmunol. 2022, 13, 42–53. [Google Scholar] [CrossRef]
- Abdellatif, B.; McVeigh, C.; Bendriss, G.; Chaari, A. The Promising Role of Probiotics in Managing the Altered Gut in Autism Spectrum Disorders. Int. J. Mol. Sci. 2020, 21, 4159. [Google Scholar] [CrossRef] [PubMed]
- Chong, H.X.; Yusoff, N.a.A.; Hor, Y.-Y.; Lew, L.-C.; Jaafar, M.H.; Choi, S.-B.; Yusoff, M.S.B.; Wahid, N.; Abdullah, M.F.I.L.; Zakaria, N.; et al. Lactobacillus plantarum DR7 alleviates stress and anxiety in adults: A randomised, double-blind, placebo-controlled study. Benef. Microbes 2019, 10, 355–373. [Google Scholar] [CrossRef]
- Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W.J. Psychobiotics and the Manipulation of Bacteria–Gut–Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef]
- Gupta, N.; Agarwal, A.K.; Al-Dossari, M.; Abd EL-Gawaad, N.S. Psychobiotic potential of Lactiplantibacillus plantarum: Current perspective in neurodegeneration and geriatric therapies. Brain Behav. Immun. Integr. 2024, 5, 100038. [Google Scholar] [CrossRef]
- Wang, W.; Liu, F.; Xu, C.; Liu, Z.; Ma, J.; Gu, L.; Jiang, Z.; Hou, J. Lactobacillus plantarum 69-2 Combined with Galacto-Oligosaccharides Alleviates d-Galactose-Induced Aging by Regulating the AMPK/SIRT1 Signaling Pathway and Gut Microbiota in Mice. J. Agric. Food Chem. 2021, 69, 2745–2757. [Google Scholar] [CrossRef]
- Yunes, R.A.; Poluektova, E.U.; Vasileva, E.V.; Odorskaya, M.V.; Marsova, M.V.; Kovalev, G.I.; Danilenko, V.N. A Multi-strain Potential Probiotic Formulation of GABA-Producing Lactobacillus plantarum 90sk and Bifidobacterium adolescentis 150 with Antidepressant Effects. Probiotics Antimicrob. Proteins 2020, 12, 973–979. [Google Scholar] [CrossRef]
- González-Bosch, C.; Boorman, E.; Zunszain, P.A.; Mann, G.E. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021, 47, 102165. [Google Scholar] [CrossRef]
- Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef]
- Tiihonen, K.; Ouwehand, A.C.; Rautonen, N. Human intestinal microbiota and healthy ageing. Ageing Res. Rev. 2010, 9, 107–116. [Google Scholar] [CrossRef] [PubMed]
- 2023 Alzheimer’s disease facts and figures. Alzheimers Dement. J. Alzheimers Assoc. 2023, 19, 1598–1695. [CrossRef]
- Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
- Asti, A.; Gioglio, L. Can a bacterial endotoxin be a key factor in the kinetics of amyloid fibril formation? J. Alzheimers Dis. JAD 2014, 39, 169–179. [Google Scholar] [CrossRef] [PubMed]
- Vom Berg, J.; Prokop, S.; Miller, K.R.; Obst, J.; Kälin, R.E.; Lopategui-Cabezas, I.; Wegner, A.; Mair, F.; Schipke, C.G.; Peters, O.; et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat. Med. 2012, 18, 1812–1819. [Google Scholar] [CrossRef]
- Schwartz, A. What’s next for Alzheimer treatment?: While Aβ isn’t out of the picture yet, several other therapeutic routes are being explored. Ann. Neurol. 2013, 73, A7–A9. [Google Scholar] [CrossRef] [PubMed]
- Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef]
- 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]
- Zhang, L.; Wang, Y.; Xiayu, X.; Shi, C.; Chen, W.; Song, N.; Fu, X.; Zhou, R.; Xu, Y.-F.; Huang, L.; et al. Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. JAD 2017, 60, 1241–1257. [Google Scholar] [CrossRef]
- Ho, L.; Ono, K.; Tsuji, M.; Mazzola, P.; Singh, R.; Pasinetti, G.M. Protective roles of intestinal microbiota derived short chain fatty acids in Alzheimer’s disease-type beta-amyloid neuropathological mechanisms. Expert Rev. Neurother. 2018, 18, 83–90. [Google Scholar] [CrossRef]
- De Rijke, T.J.; Doting, M.H.E.; van Hemert, S.; De Deyn, P.P.; Harmsen, H.J.M.; Sommer, I.E.C. A Systematic Review on the Effects of Different Types of Probiotics in Animal Alzheimer’s Disease Studies. Front. Psychiatry 2022, 13, 879491. [Google Scholar] [CrossRef] [PubMed]
- Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. JCDR 2017, 11, KC01–KC05. [Google Scholar] [CrossRef]
- Minkova, L.; Habich, A.; Peter, J.; Kaller, C.P.; Eickhoff, S.B.; Klöppel, S. Gray matter asymmetries in aging and neurodegeneration: A review and meta-analysis. Hum. Brain Mapp. 2017, 38, 5890–5904. [Google Scholar] [CrossRef] [PubMed]
- Schaub, A.-C.; Schneider, E.; Vazquez-Castellanos, J.F.; Schweinfurth, N.; Kettelhack, C.; Doll, J.P.K.; Yamanbaeva, G.; Mählmann, L.; Brand, S.; Beglinger, C.; et al. Clinical, gut microbial and neural effects of a probiotic add-on therapy in depressed patients: A randomized controlled trial. Transl. Psychiatry 2022, 12, 227. [Google Scholar] [CrossRef] [PubMed]
- Van Den Eeden, S.K.; Tanner, C.M.; Bernstein, A.L.; Fross, R.D.; Leimpeter, A.; Bloch, D.A.; Nelson, L.M. Incidence of Parkinson’s disease: Variation by age, gender, and race/ethnicity. Am. J. Epidemiol. 2003, 157, 1015–1022. [Google Scholar] [CrossRef] [PubMed]
- Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
- Lampropoulos, I.C.; Malli, F.; Sinani, O.; Gourgoulianis, K.I.; Xiromerisiou, G. Worldwide trends in mortality related to Parkinson’s disease in the period of 1994–2019: Analysis of vital registration data from the WHO Mortality Database. Front. Neurol. 2022, 13, 956440. [Google Scholar] [CrossRef]
- Schrag, A.; Horsfall, L.; Walters, K.; Noyce, A.; Petersen, I. Prediagnostic presentations of Parkinson’s disease in primary care: A case-control study. Lancet Neurol. 2015, 14, 57–64. [Google Scholar] [CrossRef]
- Balestrino, R.; Schapira, A.h.v. Parkinson disease. Eur. J. Neurol. 2020, 27, 27–42. [Google Scholar] [CrossRef]
- Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primer 2017, 3, 17013. [Google Scholar] [CrossRef] [PubMed]
- 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. Off. J. Mov. Disord. Soc. 2015, 30, 350–358. [Google Scholar] [CrossRef]
- Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef] [PubMed]
- Tan, A.H.; Chong, C.W.; Lim, S.-Y.; Yap, I.K.S.; Teh, C.S.J.; Loke, M.F.; Song, S.-L.; Tan, J.Y.; Ang, B.H.; Tan, Y.Q.; et al. Gut Microbial Ecosystem in Parkinson Disease: New Clinicobiological Insights from Multi-Omics. Ann. Neurol. 2021, 89, 546–559. [Google Scholar] [CrossRef] [PubMed]
- 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. Park. Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Aho, V.T.E.; Houser, M.C.; Pereira, P.A.B.; Chang, J.; Rudi, K.; Paulin, L.; Hertzberg, V.; Auvinen, P.; Tansey, M.G.; Scheperjans, F. Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol. Neurodegener. 2021, 16, 6. [Google Scholar] [CrossRef]
- Liu, J.; Wang, F.; Liu, S.; Du, J.; Hu, X.; Xiong, J.; Fang, R.; Chen, W.; Sun, J. Sodium butyrate exerts protective effect against Parkinson’s disease in mice via stimulation of glucagon like peptide-1. J. Neurol. Sci. 2017, 381, 176–181. [Google Scholar] [CrossRef]
- Paiva, I.; Pinho, R.; Pavlou, M.A.; Hennion, M.; Wales, P.; Schütz, A.-L.; Rajput, A.; Szego, É.M.; Kerimoglu, C.; Gerhardt, E.; et al. Sodium butyrate rescues dopaminergic cells from alpha-synuclein-induced transcriptional deregulation and DNA damage. Hum. Mol. Genet. 2017, 26, 2231–2246. [Google Scholar] [CrossRef]
- Perez Visñuk, D.; del Milagro Teran, M.; Savoy de Giori, G.; LeBlanc, J.G.; de Moreno de LeBlanc, A. Neuroprotective Effect of Riboflavin Producing Lactic Acid Bacteria in Parkinsonian Models. Neurochem. Res. 2022, 47, 1269–1279. [Google Scholar] [CrossRef]
- Tan, A.H.; Hor, J.W.; Chong, C.W.; Lim, S.-Y. Probiotics for Parkinson’s disease: Current evidence and future directions. JGH Open 2021, 5, 414–419. [Google Scholar] [CrossRef]
- Noto, D.; Miyake, S. Gut dysbiosis and multiple sclerosis. Clin. Immunol. 2022, 235, 108380. [Google Scholar] [CrossRef] [PubMed]
- Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef] [PubMed]
- Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed]
- Miyake, S.; Kim, S.; Suda, W.; Oshima, K.; Nakamura, M.; Matsuoka, T.; Chihara, N.; Tomita, A.; Sato, W.; Kim, S.-W.; et al. Dysbiosis in the Gut Microbiota of Patients with Multiple Sclerosis, with a Striking Depletion of Species Belonging to Clostridia XIVa and IV Clusters. PLoS ONE 2015, 10, e0137429. [Google Scholar] [CrossRef]
- Berer, K.; Mues, M.; Koutrolos, M.; Rasbi, Z.A.; Boziki, M.; Johner, C.; Wekerle, H.; Krishnamoorthy, G. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 2011, 479, 538–541. [Google Scholar] [CrossRef]
- Goverman, J.; Woods, A.; Larson, L.; Weiner, L.P.; Hood, L.; Zaller, D.M. Transgenic mice that express a myelin basic protein-specific T cell receptor develop spontaneous autoimmunity. Cell 1993, 72, 551–560. [Google Scholar] [CrossRef]
- Lee, Y.K.; Menezes, J.S.; Umesaki, Y.; Mazmanian, S.K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4615–4622. [Google Scholar] [CrossRef]
- Ochoa-Repáraz, J.; Mielcarz, D.W.; Haque-Begum, S.; Kasper, L.H. Induction of a regulatory B cell population in experimental allergic encephalomyelitis by alteration of the gut commensal microflora. Gut Microbes 2010, 1, 103–108. [Google Scholar] [CrossRef]
- Ochoa-Repáraz, J.; Mielcarz, D.W.; Ditrio, L.E.; Burroughs, A.R.; Foureau, D.M.; Haque-Begum, S.; Kasper, L.H. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J. Immunol. 2009, 183, 6041–6050. [Google Scholar] [CrossRef]
- Yokote, H.; Miyake, S.; Croxford, J.L.; Oki, S.; Mizusawa, H.; Yamamura, T. NKT cell-dependent amelioration of a mouse model of multiple sclerosis by altering gut flora. Am. J. Pathol. 2008, 173, 1714–1723. [Google Scholar] [CrossRef]
- Frohman, E.M.; Racke, M.K.; Raine, C.S. Multiple sclerosis—The plaque and its pathogenesis. N. Engl. J. Med. 2006, 354, 942–955. [Google Scholar] [CrossRef]
- Chen, T.; Noto, D.; Hoshino, Y.; Mizuno, M.; Miyake, S. Butyrate suppresses demyelination and enhances remyelination. J. Neuroinflammation 2019, 16, 165. [Google Scholar] [CrossRef]
- Haghikia, A.; Jörg, S.; Duscha, A.; Berg, J.; Manzel, A.; Waschbisch, A.; Hammer, A.; Lee, D.-H.; May, C.; Wilck, N.; et al. Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 2015, 43, 817–829. [Google Scholar] [CrossRef]
- Saresella, M.; Mendozzi, L.; Rossi, V.; Mazzali, F.; Piancone, F.; LaRosa, F.; Marventano, I.; Caputo, D.; Felis, G.E.; Clerici, M. Immunological and Clinical Effect of Diet Modulation of the Gut Microbiome in Multiple Sclerosis Patients: A Pilot Study. Front. Immunol. 2017, 8, 1391. [Google Scholar] [CrossRef]
- Mestre, L.; Carrillo-Salinas, F.J.; Feliú, A.; Mecha, M.; Alonso, G.; Espejo, C.; Calvo-Barreiro, L.; Luque-García, J.L.; Estevez, H.; Villar, L.M.; et al. How oral probiotics affect the severity of an experimental model of progressive multiple sclerosis? Bringing commensal bacteria into the neurodegenerative process. Gut Microbes 2020, 12, 1813532. [Google Scholar] [CrossRef] [PubMed]
- Achtert, K.; Kerkemeyer, L. The economic burden of amyotrophic lateral sclerosis: A systematic review. Eur. J. Health Econ. 2021, 22, 1151–1166. [Google Scholar] [CrossRef] [PubMed]
- Berry, J.D.; Blanchard, M.; Bonar, K.; Drane, E.; Murton, M.; Ploug, U.; Ricchetti-Masterson, K.; Savic, N.; Worthington, E.; Heiman-Patterson, T. Epidemiology and economic burden of amyotrophic lateral sclerosis in the United States: A literature review. Amyotroph. Lateral Scler. Front. Degener. 2023, 24, 436–448. [Google Scholar] [CrossRef]
- Nübling, G.S.; Mie, E.; Bauer, R.M.; Hensler, M.; Lorenzl, S.; Hapfelmeier, A.; Irwin, D.E.; Borasio, G.D.; Winkler, A.S. Increased prevalence of bladder and intestinal dysfunction in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2014, 15, 174–179. [Google Scholar] [CrossRef]
- Wu, S.; Yi, J.; Zhang, Y.-G.; Zhou, J.; Sun, J. Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model. Physiol. Rep. 2015, 3, e12356. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.-G.; Wu, S.; Yi, J.; Xia, Y.; Jin, D.; Zhou, J.; Sun, J. Target Intestinal Microbiota to Alleviate Disease Progression in Amyotrophic Lateral Sclerosis. Clin. Ther. 2017, 39, 322–336. [Google Scholar] [CrossRef] [PubMed]
- Rowin, J.; Xia, Y.; Jung, B.; Sun, J. Gut inflammation and dysbiosis in human motor neuron disease. Physiol. Rep. 2017, 5, e13443. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Baxter, A.J.; Brugha, T.S.; Erskine, H.E.; Scheurer, R.W.; Vos, T.; Scott, J.G. The epidemiology and global burden of autism spectrum disorders. Psychol. Med. 2015, 45, 601–613. [Google Scholar] [CrossRef] [PubMed]
- Lord, C.; Brugha, T.S.; Charman, T.; Cusack, J.; Dumas, G.; Frazier, T.; Jones, E.J.H.; Jones, R.M.; Pickles, A.; State, M.W.; et al. Autism spectrum disorder. Nat. Rev. Dis. Primer 2020, 6, 5. [Google Scholar] [CrossRef] [PubMed]
- Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism spectrum disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef]
- Angelis, M.D.; Piccolo, M.; Vannini, L.; Siragusa, S.; Giacomo, A.D.; Serrazzanetti, D.I.; Cristofori, F.; Guerzoni, M.E.; Gobbetti, M.; Francavilla, R. Fecal Microbiota and Metabolome of Children with Autism and Pervasive Developmental Disorder Not Otherwise Specified. PLoS ONE 2013, 8, e76993. [Google Scholar] [CrossRef]
- Tomova, A.; Husarova, V.; Lakatosova, S.; Bakos, J.; Vlkova, B.; Babinska, K.; Ostatnikova, D. Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav. 2015, 138, 179–187. [Google Scholar] [CrossRef]
- Strati, F.; Cavalieri, D.; Albanese, D.; De Felice, C.; Donati, C.; Hayek, J.; Jousson, O.; Leoncini, S.; Renzi, D.; Calabrò, A.; et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome 2017, 5, 24. [Google Scholar] [CrossRef]
- Chamtouri, M.; Merghni, A.; Gaddour, N.; Mastouri, M.; Arboleya, S.; de los Reyes-Gavilán, C.G. Lactobacilli Profile in Faecal Samples of Tunisian Children Diagnosed with Autism Spectrum Disorder. Microbiol. Res. 2023, 14, 1225–1237. [Google Scholar] [CrossRef]
- Shaaban, S.Y.; El Gendy, Y.G.; Mehanna, N.S.; El-Senousy, W.M.; El-Feki, H.S.A.; Saad, K.; El-Asheer, O.M. The role of probiotics in children with autism spectrum disorder: A prospective, open-label study. Nutr. Neurosci. 2018, 21, 676–681. [Google Scholar] [CrossRef]
- Grossi, E.; Melli, S.; Dunca, D.; Terruzzi, V. Unexpected improvement in core autism spectrum disorder symptoms after long-term treatment with probiotics. SAGE Open Med. Case Rep. 2016, 4, 2050313X16666231. [Google Scholar] [CrossRef]
- Foss-Feig, J.H.; Adkinson, B.D.; Ji, J.L.; Yang, G.; Srihari, V.H.; McPartland, J.C.; Krystal, J.H.; Murray, J.D.; Anticevic, A. Searching for Cross-Diagnostic Convergence: Neural Mechanisms Governing Excitation and Inhibition Balance in Schizophrenia and Autism Spectrum Disorders. Biol. Psychiatry 2017, 81, 848–861. [Google Scholar] [CrossRef] [PubMed]
- Kong, Q.; Chen, Q.; Mao, X.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium longum CCFM1077 Ameliorated Neurotransmitter Disorder and Neuroinflammation Closely Linked to Regulation in the Kynurenine Pathway of Autistic-like Rats. Nutrients 2022, 14, 1615. [Google Scholar] [CrossRef]
- Zhang, Y.; Guo, M.; Zhang, H.; Wang, Y.; Li, R.; Liu, Z.; Zheng, H.; You, C. Lactiplantibacillus plantarum ST-III-fermented milk improves autistic-like behaviors in valproic acid-induced autism spectrum disorder mice by altering gut microbiota. Front. Nutr. 2022, 9, 1005308. [Google Scholar] [CrossRef]
- Mehra, A.; Arora, G.; Sahni, G.; Kaur, M.; Singh, H.; Singh, B.; Kaur, S. Gut microbiota and Autism Spectrum Disorder: From pathogenesis to potential therapeutic perspectives. J. Tradit. Complement. Med. 2023, 13, 135–149. [Google Scholar] [CrossRef]
- Kong, X.-J.; Kang, J.; Liu, K. Probiotic and intra-nasal oxytocin combination therapy on autonomic function and gut-brain axis signaling in young children and teens with autism spectrum disorder. J. Psychiatr. Res. 2023, 166, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
- Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H.; et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef] [PubMed]
- Vich Vila, A.; Collij, V.; Sanna, S.; Sinha, T.; Imhann, F.; Bourgonje, A.R.; Mujagic, Z.; Jonkers, D.M.A.E.; Masclee, A.A.M.; Fu, J.; et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 2020, 11, 362. [Google Scholar] [CrossRef]
- Zeng, C.; Yang, P.; Cao, T.; Gu, Y.; Li, N.; Zhang, B.; Xu, P.; Liu, Y.; Luo, Z.; Cai, H. Gut microbiota: An intermediary between metabolic syndrome and cognitive deficits in schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 106, 110097. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Wang, K.; Hu, J. Effect of Probiotics on Depression: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2016, 8, 483. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.J.; Hong, J.K.; Kim, J.-K.; Kim, D.-H.; Jang, S.W.; Han, S.-W.; Yoon, I.-Y. Effects of Probiotic NVP-1704 on Mental Health and Sleep in Healthy Adults: An 8-Week Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2021, 13, 2660. [Google Scholar] [CrossRef]
- Ayichew, T.; Belete, A.; Alebachew, T.; Tsehaye, H.; Berhanu, H.; Minwuyelet, A. Bacterial Probiotics their Importances and Limitations: A Review. J. Nutr. Health Sci. 2017, 4, 202. [Google Scholar] [CrossRef]
- Ferstl, R.; Frei, R.; Schiavi, E.; Konieczna, P.; Barcik, W.; Ziegler, M.; Lauener, R.P.; Chassard, C.; Lacroix, C.; Akdis, C.A.; et al. Histamine receptor 2 is a key influence in immune responses to intestinal histamine-secreting microbes. J. Allergy Clin. Immunol. 2014, 134, 744–746.e3. [Google Scholar] [CrossRef] [PubMed]
- Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R.B.-Z.; et al. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018, 174, 1388–1405.e21. [Google Scholar] [CrossRef] [PubMed]
- Arora, K.; Green, M.; Prakash, S. The Microbiome and Alzheimer’s Disease: Potential and Limitations of Prebiotic, Synbiotic, and Probiotic Formulations. Front. Bioeng. Biotechnol. 2020, 8, 537847. [Google Scholar] [CrossRef] [PubMed]
Alzheimer’s Disease | Parkinson’s Disease | Multiple Sclerosis | Amyotrophic Lateral Sclerosis | Autism Spectrum Disorder | Anxiety and Depression |
---|---|---|---|---|---|
Lactiplantibacillus plantarum R1012 [108] | Lactiplantibacillus plantarum PS128 [109] | Lactiplantibacillus plantarum DSM 24730 [110] | Lactiplantibacillus acidophilus [111] | Lactiplantibacillus plantarum ST-III [112] | Lactiplantibacillus plantarum strain PS128 [113] |
Bifidobacterium longum NK46 [114] | Lactiplantibacillus plantarum DP189 [115] | Bifidobacterium longum DSM 2436 [110] | Lactobacillus fermentum [116] | Lactiplantibacillus plantarum PS128 [117] | Lactiplantibacillus plantarum APsulloc 331261 [118] |
Lactobacillus helveticus IDCC3801 [119] | Lactiplantibacillus plantarum NCIMB 30173 [120] | Streptococcus thermophilus DSM 2431 [110] | Lactobacillus delbrueckii [116] | Lactiplantibacillus plantarum WCFS1 [121] | Lactiplantibacillus plantarum 299v [107] |
Lactiplantibacillus plantarum MTCC1325- [122] | Lactobacillus rhamnosus NCIMB 30174 [111] | Bifidobacterium Animalis PTCC 1631 [123] | Lactobacillus salivarius [116] | Bifidobacterium lactis Probio-M8 [124] | Lactiplantibacillus plantarum JYLP-326 [125] |
Lactiplantibacillus plantarum DSM 32244 [126] | Lactobacillus acidophilus NCIMB 30175 [120] | Lactiplantibacillus plantarum 1058 [127] | Bifidobacterium [116] | Lactobacillus rhamnosus HN001 [128] | Lactiplantibacillus plantarum DR7 [129] |
Lactiplantibacillus Strain | Neurodegenerative Disease | Protocol Summary |
---|---|---|
Lactiplantibacillus plantarum PS128 | Parkinson Disease | n participants = 25 Duration of treatment = 12 weeks |
Lactiplantibacillus plantarum 299v [107] | Anxiety and Depression | n participants = 40 Duration of treatment = 8 weeks |
Lactiplantibacillus acidophilus | Amyotrophic Lateral Sclerosis | n participants = 42 Duration of treatment = 12 weeks |
Lactiplantibacillus plantarum DSM 24730 | Multiple Sclerosis | n participants = 9 Duration of treatment = 2 months |
Lactiplantibacillus plantarum NCIMB 30173 | Parkinson Disease | n participants = 3 Duration of treatment = 48 h |
Lactiplantibacillus plantarum MTCC1325 | Alzheimer’s Disease | n participants = 3 Duration of treatment = 48 h |
Lactiplantibacillus plantarum WCFS1 [200] | Autism Spectrum Disorder | n participants = 35 |
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. |
© 2024 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
Di Chiano, M.; Sallustio, F.; Fiocco, D.; Rocchetti, M.T.; Spano, G.; Pontrelli, P.; Moschetta, A.; Gesualdo, L.; Gadaleta, R.M.; Gallone, A. Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases. Int. J. Mol. Sci. 2024, 25, 9489. https://doi.org/10.3390/ijms25179489
Di Chiano M, Sallustio F, Fiocco D, Rocchetti MT, Spano G, Pontrelli P, Moschetta A, Gesualdo L, Gadaleta RM, Gallone A. Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2024; 25(17):9489. https://doi.org/10.3390/ijms25179489
Chicago/Turabian StyleDi Chiano, Mariagiovanna, Fabio Sallustio, Daniela Fiocco, Maria Teresa Rocchetti, Giuseppe Spano, Paola Pontrelli, Antonio Moschetta, Loreto Gesualdo, Raffaella Maria Gadaleta, and Anna Gallone. 2024. "Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases" International Journal of Molecular Sciences 25, no. 17: 9489. https://doi.org/10.3390/ijms25179489
APA StyleDi Chiano, M., Sallustio, F., Fiocco, D., Rocchetti, M. T., Spano, G., Pontrelli, P., Moschetta, A., Gesualdo, L., Gadaleta, R. M., & Gallone, A. (2024). Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases. International Journal of Molecular Sciences, 25(17), 9489. https://doi.org/10.3390/ijms25179489