Skip to main content
Top
Published in: Journal of Translational Medicine 1/2022

Open Access 01-12-2022 | Probiotics | Review

Recent developments in the probiotics as live biotherapeutic products (LBPs) as modulators of gut brain axis related neurological conditions

Authors: Duygu Ağagündüz, Feray Gençer Bingöl, Elif Çelik, Özge Cemali, Çiler Özenir, Fatih Özoğul, Raffaele Capasso

Published in: Journal of Translational Medicine | Issue 1/2022

Login to get access

Abstract

Probiotics have been defined as “living microorganisms that create health benefits in the host when taken in sufficient amounts. Recent developments in the understanding of the relationship between the microbiom and its host have shown evidence about the promising potential of probiotics to improve certain health problems. However, today, there are some confusions about traditional and new generation foods containing probiotics, naming and classifications of them in scientific studies and also their marketing. To clarify this confusion, the Food and Drug Administration (FDA) declared that it has made a new category definition called "live biotherapeutic products" (LBPs). Accordingly, the FDA has designated LBPs as “a biological product that: i)contains live organisms, such as bacteria; ii)is applicable to the prevention, treatment, or cure of a disease/condition of human beings; and iii) is not a vaccine”. The accumulated literature focused on LBPs to determine effective strains in health and disease, and often focused on obesity, diabetes, and certain diseases like inflammatory bowel disease (IBD).However, microbiome also play an important role in the pathogenesis of diseases that age day by day in the modern world via gut-brain axis. Herein, we discuss the novel roles of LBPs in some gut-brain axis related conditions in the light of recent studies. This article may be of interest to a broad readership including those interested in probiotics as LBPs, their health effects and safety, also gut-brain axis.
Literature
6.
go back to reference Berg G, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8(1):1–22. Berg G, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8(1):1–22.
9.
10.
go back to reference Pham VT, et al. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res. 2021;95:35–53.PubMedCrossRef Pham VT, et al. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res. 2021;95:35–53.PubMedCrossRef
11.
go back to reference Singh A, et al. Interaction of polyphenols as antioxidant and anti-ınflammatory compounds in brain-liver-gut axis. Antioxidants. 2020;9(8):669.PubMedCentralCrossRef Singh A, et al. Interaction of polyphenols as antioxidant and anti-ınflammatory compounds in brain-liver-gut axis. Antioxidants. 2020;9(8):669.PubMedCentralCrossRef
12.
go back to reference Cordaillat-Simmons M, Rouanet A, Pot B. Live biotherapeutic products: the importance of a defined regulatory framework. Exp Mol Med. 2020;52(9):1397–406.PubMedPubMedCentralCrossRef Cordaillat-Simmons M, Rouanet A, Pot B. Live biotherapeutic products: the importance of a defined regulatory framework. Exp Mol Med. 2020;52(9):1397–406.PubMedPubMedCentralCrossRef
13.
go back to reference FDA, center for biologics evaluation and research. Early clinical trials with live biotherapeutic products: chemistry,manufacturing, and control ınformation 2016. FDA, center for biologics evaluation and research. Early clinical trials with live biotherapeutic products: chemistry,manufacturing, and control ınformation 2016.
14.
go back to reference Pharmacopoeia, E., 3053E General monograph on live biotherapeutic products. European pharmacopoeia 9 7 2019. Pharmacopoeia, E., 3053E General monograph on live biotherapeutic products. European pharmacopoeia 9 7 2019.
16.
go back to reference Kazemi A, et al. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: a randomized clinical trial. Clin Nutr. 2019;38(2):522–8.PubMedCrossRef Kazemi A, et al. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: a randomized clinical trial. Clin Nutr. 2019;38(2):522–8.PubMedCrossRef
17.
go back to reference Wu SI, et al. Psychobiotic supplementation of PS128(TM) improves stress, anxiety, and insomnia in highly stressed information technology specialists: a pilot study. Front Nutr. 2021;8:614105.PubMedPubMedCentralCrossRef Wu SI, et al. Psychobiotic supplementation of PS128(TM) improves stress, anxiety, and insomnia in highly stressed information technology specialists: a pilot study. Front Nutr. 2021;8:614105.PubMedPubMedCentralCrossRef
18.
go back to reference Shahrbabaki ME, et al. The efficacy of probiotics for treatment of bipolar disorder-type 1: a randomized, double-blind, placebo controlled trial. Iran J Psychiatry. 2020;15(1):10. Shahrbabaki ME, et al. The efficacy of probiotics for treatment of bipolar disorder-type 1: a randomized, double-blind, placebo controlled trial. Iran J Psychiatry. 2020;15(1):10.
19.
go back to reference Yamamura R, et al. Lipid and energy metabolism of the gut microbiota is associated with the response to probiotic Bifidobacterium breve strain for anxiety and depressive symptoms in schizophrenia. J Pers Med. 2021;11(10):987.PubMedPubMedCentralCrossRef Yamamura R, et al. Lipid and energy metabolism of the gut microbiota is associated with the response to probiotic Bifidobacterium breve strain for anxiety and depressive symptoms in schizophrenia. J Pers Med. 2021;11(10):987.PubMedPubMedCentralCrossRef
20.
go back to reference Mörkl S, et al. Probiotics and the microbiota-gut-brain axis: focus on psychiatry. Current Nutr Rep. 2020;9(3):171–82.CrossRef Mörkl S, et al. Probiotics and the microbiota-gut-brain axis: focus on psychiatry. Current Nutr Rep. 2020;9(3):171–82.CrossRef
21.
go back to reference Umbrello G, Esposito S. Microbiota and neurologic diseases: potential effects of probiotics. J Transl Med. 2016;14(1):1–11.CrossRef Umbrello G, Esposito S. Microbiota and neurologic diseases: potential effects of probiotics. J Transl Med. 2016;14(1):1–11.CrossRef
22.
go back to reference Association AP. Diagnostic and statistical manual of mental health disorders, (DSM-5). Washington DC: American Psychiatric Publishing; 2013.CrossRef Association AP. Diagnostic and statistical manual of mental health disorders, (DSM-5). Washington DC: American Psychiatric Publishing; 2013.CrossRef
23.
go back to reference Davies C, et al. Altering the gut microbiome to potentially modulate behavioral manifestations in autism spectrum disorders: a systematic review. Neurosci Biobehav Rev. 2021;128:549–57.PubMedCrossRef Davies C, et al. Altering the gut microbiome to potentially modulate behavioral manifestations in autism spectrum disorders: a systematic review. Neurosci Biobehav Rev. 2021;128:549–57.PubMedCrossRef
24.
go back to reference Crawford S. On the origins of autism: the quantitative threshold exposure hypothesis. Med Hypotheses. 2015;85(6):798–806.PubMedCrossRef Crawford S. On the origins of autism: the quantitative threshold exposure hypothesis. Med Hypotheses. 2015;85(6):798–806.PubMedCrossRef
25.
go back to reference Geier DA, et al. A prospective study of transsulfuration biomarkers in autistic disorders. Neurochem Res. 2009;34(2):386–93.PubMedCrossRef Geier DA, et al. A prospective study of transsulfuration biomarkers in autistic disorders. Neurochem Res. 2009;34(2):386–93.PubMedCrossRef
26.
go back to reference Snigdha S, et al. Probiotics: potential novel therapeutics for microbiota-gut-brain axis dysfunction across gender and lifespan. Pharmacol Therapeutics. 2021;231:107978.CrossRef Snigdha S, et al. Probiotics: potential novel therapeutics for microbiota-gut-brain axis dysfunction across gender and lifespan. Pharmacol Therapeutics. 2021;231:107978.CrossRef
29.
go back to reference Kałużna-Czaplińska J, Jóźwik-Pruska J. Nutritional strategies and personalized diet in autism-choice or necessity? Trends Food Sci Technol. 2016;49:45–50.CrossRef Kałużna-Czaplińska J, Jóźwik-Pruska J. Nutritional strategies and personalized diet in autism-choice or necessity? Trends Food Sci Technol. 2016;49:45–50.CrossRef
30.
go back to reference Horvath K, Perman JA. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002;4(3):251–8.PubMedCrossRef Horvath K, Perman JA. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002;4(3):251–8.PubMedCrossRef
31.
go back to reference de Magistris L, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418–24.PubMedCrossRef de Magistris L, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418–24.PubMedCrossRef
32.
go back to reference Heberling CA, Dhurjati PS, Sasser M. Hypothesis for a systems connectivity model of autism spectrum disorder pathogenesis: links to gut bacteria, oxidative stress, and intestinal permeability. Med Hypotheses. 2013;80(3):264–70.PubMedCrossRef Heberling CA, Dhurjati PS, Sasser M. Hypothesis for a systems connectivity model of autism spectrum disorder pathogenesis: links to gut bacteria, oxidative stress, and intestinal permeability. Med Hypotheses. 2013;80(3):264–70.PubMedCrossRef
33.
go back to reference Finegold SM, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16(4):444–53.PubMedCrossRef Finegold SM, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16(4):444–53.PubMedCrossRef
34.
go back to reference Berding K, Donovan SM. Microbiome and nutrition in autism spectrum disorder: current knowledge and research needs. Nutr Rev. 2016;74(12):723–36.PubMedCrossRef Berding K, Donovan SM. Microbiome and nutrition in autism spectrum disorder: current knowledge and research needs. Nutr Rev. 2016;74(12):723–36.PubMedCrossRef
35.
go back to reference Adams JB, et al. Gastrointestinal flora and gastrointestinal status in children with autism–comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011;11(1):1–13.CrossRef Adams JB, et al. Gastrointestinal flora and gastrointestinal status in children with autism–comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011;11(1):1–13.CrossRef
36.
go back to reference Wang L, et al. Increased abundance of Sutterella spp. and ruminococcus torques in feces of children with autism spectrum disorder. Mol Autism. 2013;4(1):1–4.CrossRef Wang L, et al. Increased abundance of Sutterella spp. and ruminococcus torques in feces of children with autism spectrum disorder. Mol Autism. 2013;4(1):1–4.CrossRef
37.
go back to reference Parracho HM, et al. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J Med Microbiol. 2005;54(10):987–91.PubMedCrossRef Parracho HM, et al. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J Med Microbiol. 2005;54(10):987–91.PubMedCrossRef
38.
go back to reference Tomova A, et al. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. 2015;138:179–87.PubMedCrossRef Tomova A, et al. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. 2015;138:179–87.PubMedCrossRef
39.
40.
go back to reference Maiuolo J, et al. The contribution of gut microbiota-brain axis in the development of brain disorders. Front Neurosci. 2021;15:170.CrossRef Maiuolo J, et al. The contribution of gut microbiota-brain axis in the development of brain disorders. Front Neurosci. 2021;15:170.CrossRef
41.
go back to reference Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009;6(5):306–14.PubMedCrossRef Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009;6(5):306–14.PubMedCrossRef
42.
go back to reference Naveed M, et al. Gut-brain axis: A matter of concern in neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2021;104: 110051.PubMedCrossRef Naveed M, et al. Gut-brain axis: A matter of concern in neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2021;104: 110051.PubMedCrossRef
44.
go back to reference Pangrazzi L, Balasco L, Bozzi Y. Oxidative stress and ımmune system dysfunction in autism spectrum disorders. Int J Mol Sci. 2020;21(9):3293.PubMedCentralCrossRef Pangrazzi L, Balasco L, Bozzi Y. Oxidative stress and ımmune system dysfunction in autism spectrum disorders. Int J Mol Sci. 2020;21(9):3293.PubMedCentralCrossRef
45.
go back to reference Swann JR, Spitzer SO, Diaz Heijtz R. Developmental signatures of microbiota-derived metabolites in the mouse brain. Metabolites. 2020;10(5):172.PubMedCentralCrossRef Swann JR, Spitzer SO, Diaz Heijtz R. Developmental signatures of microbiota-derived metabolites in the mouse brain. Metabolites. 2020;10(5):172.PubMedCentralCrossRef
48.
go back to reference Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):919–29.PubMedCrossRef Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):919–29.PubMedCrossRef
49.
go back to reference MacFabe DF. Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microb Ecol Health Dis. 2012;23(1):19260. MacFabe DF. Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microb Ecol Health Dis. 2012;23(1):19260.
50.
go back to reference Dowhaniuk JK, et al. Starving the gut: a deficit of butyrate in the intestinal ecosystem of children with intestinal failure. J Parenter Enter Nutr. 2020;44(6):1112–23.CrossRef Dowhaniuk JK, et al. Starving the gut: a deficit of butyrate in the intestinal ecosystem of children with intestinal failure. J Parenter Enter Nutr. 2020;44(6):1112–23.CrossRef
51.
go back to reference Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol. 2020;11:25.CrossRef Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol. 2020;11:25.CrossRef
52.
go back to reference Wenzel TJ, et al. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Mol Cell Neurosci. 2020;105:103493.PubMedCrossRef Wenzel TJ, et al. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Mol Cell Neurosci. 2020;105:103493.PubMedCrossRef
54.
go back to reference Sharpley CF, et al. Further evidence of HPA-axis dysregulation and its correlation with depression in autism spectrum disorders: data from girls. Physiol Behav. 2016;167:110–7.PubMedCrossRef Sharpley CF, et al. Further evidence of HPA-axis dysregulation and its correlation with depression in autism spectrum disorders: data from girls. Physiol Behav. 2016;167:110–7.PubMedCrossRef
55.
go back to reference Kushki A, et al. Functional autonomic nervous system profile in children with autism spectrum disorder. Molecular Autism. 2014;5(1):1–10.CrossRef Kushki A, et al. Functional autonomic nervous system profile in children with autism spectrum disorder. Molecular Autism. 2014;5(1):1–10.CrossRef
56.
go back to reference Sivamaruthi BS, et al. The role of microbiome, dietary supplements, and probiotics in autism spectrum disorder. Int J Environ Res Public Health. 2020;17(8):2647.PubMedCentralCrossRef Sivamaruthi BS, et al. The role of microbiome, dietary supplements, and probiotics in autism spectrum disorder. Int J Environ Res Public Health. 2020;17(8):2647.PubMedCentralCrossRef
57.
go back to reference Li Q, Zhou J-M. The microbiota–gut–brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience. 2016;324:131–9.PubMedCrossRef Li Q, Zhou J-M. The microbiota–gut–brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience. 2016;324:131–9.PubMedCrossRef
58.
go back to reference Pochakom A, et al. Selective probiotic treatment positively modulates the microbiota-gut-brain axis in the btbr mouse model of autism. Brain Sci. 2022;12(6):781.PubMedPubMedCentralCrossRef Pochakom A, et al. Selective probiotic treatment positively modulates the microbiota-gut-brain axis in the btbr mouse model of autism. Brain Sci. 2022;12(6):781.PubMedPubMedCentralCrossRef
59.
go back to reference Tabouy L, et al. Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain Behav Immun. 2018;73:310–9.PubMedCrossRef Tabouy L, et al. Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain Behav Immun. 2018;73:310–9.PubMedCrossRef
60.
go back to reference Bravo JA, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci. 2011;108(38):16050–5.PubMedPubMedCentralCrossRef Bravo JA, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci. 2011;108(38):16050–5.PubMedPubMedCentralCrossRef
61.
go back to reference Liu Y-W, et al. Effects of Lactobacillus plantarum PS128 on children with autism spectrum disorder in Taiwan: a randomized, double-blind, placebo-controlled trial. Nutrients. 2019;11(4):820.PubMedCentralCrossRef Liu Y-W, et al. Effects of Lactobacillus plantarum PS128 on children with autism spectrum disorder in Taiwan: a randomized, double-blind, placebo-controlled trial. Nutrients. 2019;11(4):820.PubMedCentralCrossRef
62.
go back to reference Arnold LE, et al. Probiotics for gastrointestinal symptoms and quality of life in autism: a placebo-controlled pilot trial. J Child Adolesc Psychopharmacol. 2019;29(9):659–69.PubMedPubMedCentralCrossRef Arnold LE, et al. Probiotics for gastrointestinal symptoms and quality of life in autism: a placebo-controlled pilot trial. J Child Adolesc Psychopharmacol. 2019;29(9):659–69.PubMedPubMedCentralCrossRef
63.
go back to reference Shaaban SY, et al. The role of probiotics in children with autism spectrum disorder: a prospective, open-label study. Nutr Neurosci. 2018;21(9):676–81.PubMedCrossRef Shaaban SY, et al. The role of probiotics in children with autism spectrum disorder: a prospective, open-label study. Nutr Neurosci. 2018;21(9):676–81.PubMedCrossRef
64.
go back to reference Santocchi E, et al. Effects of probiotic supplementation on gastrointestinal, sensory and core symptoms in autism spectrum disorders: a randomized controlled trial. Front Psychiatry. 2020;2020(11):550593.CrossRef Santocchi E, et al. Effects of probiotic supplementation on gastrointestinal, sensory and core symptoms in autism spectrum disorders: a randomized controlled trial. Front Psychiatry. 2020;2020(11):550593.CrossRef
65.
go back to reference Leblhuber F, et al. Probiotic supplementation in patients with Alzheimer’s dementia-an explorative intervention study. Curr Alzheimer Res. 2018;15(12):1106–13.PubMedPubMedCentralCrossRef Leblhuber F, et al. Probiotic supplementation in patients with Alzheimer’s dementia-an explorative intervention study. Curr Alzheimer Res. 2018;15(12):1106–13.PubMedPubMedCentralCrossRef
66.
go back to reference Agahi A, et al. Does severity of Alzheimer’s disease contribute to its responsiveness to modifying gut microbiota? a double blind clinical trial. Front Neurol. 2018;9:662–662.PubMedPubMedCentralCrossRef Agahi A, et al. Does severity of Alzheimer’s disease contribute to its responsiveness to modifying gut microbiota? a double blind clinical trial. Front Neurol. 2018;9:662–662.PubMedPubMedCentralCrossRef
68.
go back to reference Sanborn V, et al. Randomized clinical trial examining the impact of Lactobacillus rhamnosus GG probiotic supplementation on cognitive functioning in middle-aged and older adults. Neuropsychiatr Dis Treat. 2020;16:2765–77.PubMedPubMedCentralCrossRef Sanborn V, et al. Randomized clinical trial examining the impact of Lactobacillus rhamnosus GG probiotic supplementation on cognitive functioning in middle-aged and older adults. Neuropsychiatr Dis Treat. 2020;16:2765–77.PubMedPubMedCentralCrossRef
69.
go back to reference Tamtaji OR, et al. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38(3):1031–5.PubMedCrossRef Tamtaji OR, et al. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38(3):1031–5.PubMedCrossRef
70.
go back to reference Tan AH, et al. Probiotics for constipation in Parkinson disease: a randomized placebo-controlled study. Neurology. 2021;96(5):e772–82.PubMed Tan AH, et al. Probiotics for constipation in Parkinson disease: a randomized placebo-controlled study. Neurology. 2021;96(5):e772–82.PubMed
72.
73.
go back to reference Azm AS, et al. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-amyloid (1–42) injected rats. Appl Physiol Nutr Metab. 2018;43(7):718–26.CrossRef Azm AS, et al. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-amyloid (1–42) injected rats. Appl Physiol Nutr Metab. 2018;43(7):718–26.CrossRef
74.
go back to reference Kaur H, et al. Effects of probiotic supplementation on short chain fatty acids in the App NL-GF mouse model of Alzheimer’s disease. J Alzheimer’s Dis. 2020;76(3):1–20. Kaur H, et al. Effects of probiotic supplementation on short chain fatty acids in the App NL-GF mouse model of Alzheimer’s disease. J Alzheimer’s Dis. 2020;76(3):1–20.
75.
go back to reference Shamsipour S, Sharifi G, Taghian F. Impact of interval training with probiotic (L plantarum/Bifidobacterium bifidum) on passive avoidance test, ChAT and BDNF in the hippocampus of rats with Alzheimer’s disease. Neurosci Lett. 2021;756:135949.PubMedCrossRef Shamsipour S, Sharifi G, Taghian F. Impact of interval training with probiotic (L plantarum/Bifidobacterium bifidum) on passive avoidance test, ChAT and BDNF in the hippocampus of rats with Alzheimer’s disease. Neurosci Lett. 2021;756:135949.PubMedCrossRef
76.
go back to reference Rezaeiasl Z, Salami M, Sepehri G. The effects of probiotic Lactobacillus and Bifidobacterium strains on memory and learning behavior, long-term potentiation (LTP), and some biochemical parameters in β-amyloid-induced rat’s model of Alzheimer’s disease. Preventive Nutrition Food Sci. 2019;24(3):265.CrossRef Rezaeiasl Z, Salami M, Sepehri G. The effects of probiotic Lactobacillus and Bifidobacterium strains on memory and learning behavior, long-term potentiation (LTP), and some biochemical parameters in β-amyloid-induced rat’s model of Alzheimer’s disease. Preventive Nutrition Food Sci. 2019;24(3):265.CrossRef
78.
go back to reference Ou Z, et al. Protective effects of Akkermansia muciniphila on cognitive deficits and amyloid pathology in a mouse model of Alzheimer’s disease. Nutr Diabetes. 2020;10(1):1–10.CrossRef Ou Z, et al. Protective effects of Akkermansia muciniphila on cognitive deficits and amyloid pathology in a mouse model of Alzheimer’s disease. Nutr Diabetes. 2020;10(1):1–10.CrossRef
79.
go back to reference Zhu G, et al. Administration of bifidobacterium breve improves the brain function of Aβ(1–42)-treated mice via the modulation of the gut microbiome. Nutrients. 2021;13(5):1602.PubMedPubMedCentralCrossRef Zhu G, et al. Administration of bifidobacterium breve improves the brain function of Aβ(1–42)-treated mice via the modulation of the gut microbiome. Nutrients. 2021;13(5):1602.PubMedPubMedCentralCrossRef
80.
go back to reference Sun J, et al. Probiotic Clostridium butyricum ameliorated motor deficits in a mouse model of Parkinson’s disease via gut microbiota-GLP-1 pathway. Brain Behav Immun. 2021;91:703–15.PubMedCrossRef Sun J, et al. Probiotic Clostridium butyricum ameliorated motor deficits in a mouse model of Parkinson’s disease via gut microbiota-GLP-1 pathway. Brain Behav Immun. 2021;91:703–15.PubMedCrossRef
81.
go back to reference Alipour Nosrani E, et al. Neuroprotective effects of probiotics bacteria on animal model of Parkinson’s disease induced by 6-hydroxydopamine: a behavioral, biochemical, and histological study. J Immunoassay Immunochem. 2021;42(2):106–20.PubMedCrossRef Alipour Nosrani E, et al. Neuroprotective effects of probiotics bacteria on animal model of Parkinson’s disease induced by 6-hydroxydopamine: a behavioral, biochemical, and histological study. J Immunoassay Immunochem. 2021;42(2):106–20.PubMedCrossRef
82.
go back to reference Tsao S-P, et al. Probiotic enhancement of antioxidant capacity and alterations of gut microbiota composition in 6-hydroxydopamin-induced parkinson’s disease rats. Antioxidants. 2021;10(11):1823.PubMedPubMedCentralCrossRef Tsao S-P, et al. Probiotic enhancement of antioxidant capacity and alterations of gut microbiota composition in 6-hydroxydopamin-induced parkinson’s disease rats. Antioxidants. 2021;10(11):1823.PubMedPubMedCentralCrossRef
83.
go back to reference Visñuk DP, et al. Neuroprotective effects associated with immune modulation by selected lactic acid bacteria in a Parkinson’s disease model. Nutrition. 2020;79:110995.CrossRef Visñuk DP, et al. Neuroprotective effects associated with immune modulation by selected lactic acid bacteria in a Parkinson’s disease model. Nutrition. 2020;79:110995.CrossRef
85.
go back to reference Ishii T, et al. Oral administration of probiotic bifidobacterium breve improves facilitation of hippocampal memory extinction via restoration of aberrant higher induction of neuropsin in an MPTP-induced mouse model of Parkinson’s disease. Biomedicines. 2021;9(2):167.PubMedPubMedCentralCrossRef Ishii T, et al. Oral administration of probiotic bifidobacterium breve improves facilitation of hippocampal memory extinction via restoration of aberrant higher induction of neuropsin in an MPTP-induced mouse model of Parkinson’s disease. Biomedicines. 2021;9(2):167.PubMedPubMedCentralCrossRef
86.
go back to reference Xie C, Prasad AA. Probiotics treatment improves hippocampal dependent cognition in a rodent model of Parkinson’s disease. Microorganisms. 2020;8(11):1661.PubMedCentralCrossRef Xie C, Prasad AA. Probiotics treatment improves hippocampal dependent cognition in a rodent model of Parkinson’s disease. Microorganisms. 2020;8(11):1661.PubMedCentralCrossRef
87.
go back to reference Khan S, Barve KH, Kumar MS. Recent advancements in pathogenesis, diagnostics and treatment of Alzheimer’s disease. Curr Neuropharmacol. 2020;18(11):1106–25.PubMedPubMedCentralCrossRef Khan S, Barve KH, Kumar MS. Recent advancements in pathogenesis, diagnostics and treatment of Alzheimer’s disease. Curr Neuropharmacol. 2020;18(11):1106–25.PubMedPubMedCentralCrossRef
88.
go back to reference Alzheimer’s Association. 2021 Alzheimer’s disease facts and figures. Alzheimers Dement 2021;17(3). Alzheimer’s Association. 2021 Alzheimer’s disease facts and figures. Alzheimers Dement 2021;17(3).
89.
go back to reference Gauthier S, Rosa-Neto P, Morais JA, Webster C. World Alzheimer Report 2021: Journey through the diagnosis of dementia. London, England: Alzheimer’s Disease International; 2021. Gauthier S, Rosa-Neto P, Morais JA, Webster C. World Alzheimer Report 2021: Journey through the diagnosis of dementia. London, England: Alzheimer’s Disease International; 2021.
90.
go back to reference Watamura N, et al. Colocalization of phosphorylated forms of WAVE1, CRMP2, and tau in Alzheimer’s disease model mice: Involvement of Cdk5 phosphorylation and the effect of ATRA treatment. J Neurosci Res. 2016;94(1):15–26.PubMedCrossRef Watamura N, et al. Colocalization of phosphorylated forms of WAVE1, CRMP2, and tau in Alzheimer’s disease model mice: Involvement of Cdk5 phosphorylation and the effect of ATRA treatment. J Neurosci Res. 2016;94(1):15–26.PubMedCrossRef
91.
92.
93.
go back to reference Cioffi F, Adam RHI, Broersen K. Molecular mechanisms and genetics of oxidative stress in Alzheimer’s disease. J Alzheimer’s Dis. 2019;72(4):981–1017.CrossRef Cioffi F, Adam RHI, Broersen K. Molecular mechanisms and genetics of oxidative stress in Alzheimer’s disease. J Alzheimer’s Dis. 2019;72(4):981–1017.CrossRef
94.
95.
96.
go back to reference Shen XN, et al. Inflammatory markers in Alzheimer’s disease and mild cognitive impairment: a meta-analysis and systematic review of 170 studies. J Neurol Neurosurg Psychiatry. 2019;90(5):590–8.PubMedCrossRef Shen XN, et al. Inflammatory markers in Alzheimer’s disease and mild cognitive impairment: a meta-analysis and systematic review of 170 studies. J Neurol Neurosurg Psychiatry. 2019;90(5):590–8.PubMedCrossRef
97.
go back to reference Chen X, et al. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9:2122.PubMedPubMedCentralCrossRef Chen X, et al. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9:2122.PubMedPubMedCentralCrossRef
98.
go back to reference Ismael S, et al. Renin-angiotensin system alterations in the human Alzheimer’s disease brain. J Alzheimers Dis. 2021;84(4):1473–84.PubMedCrossRef Ismael S, et al. Renin-angiotensin system alterations in the human Alzheimer’s disease brain. J Alzheimers Dis. 2021;84(4):1473–84.PubMedCrossRef
99.
go back to reference Scotti L, et al. Association between renin-angiotensin-aldosterone system inhibitors and risk of dementia: a meta-analysis. Pharmacol Res. 2021;166:105515.PubMedCrossRef Scotti L, et al. Association between renin-angiotensin-aldosterone system inhibitors and risk of dementia: a meta-analysis. Pharmacol Res. 2021;166:105515.PubMedCrossRef
102.
go back to reference Ribeiro VT, et al. Circulating angiotensin-(1–7) is reduced in alzheimer’s disease patients and correlates with white matter abnormalities: results from a pilot study. Front Neurosci. 2021;15:636754.PubMedPubMedCentralCrossRef Ribeiro VT, et al. Circulating angiotensin-(1–7) is reduced in alzheimer’s disease patients and correlates with white matter abnormalities: results from a pilot study. Front Neurosci. 2021;15:636754.PubMedPubMedCentralCrossRef
103.
105.
go back to reference Saji N, et al. Analysis of the relationship between the gut microbiome and dementia: a cross-sectional study conducted in Japan. Sci Rep. 2019;9(1):1008–1008.PubMedPubMedCentralCrossRef Saji N, et al. Analysis of the relationship between the gut microbiome and dementia: a cross-sectional study conducted in Japan. Sci Rep. 2019;9(1):1008–1008.PubMedPubMedCentralCrossRef
106.
go back to reference Li B, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019;15(10):1357–66.PubMedCrossRef Li B, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019;15(10):1357–66.PubMedCrossRef
107.
go back to reference Marizzoni M, et al. Short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer’s disease. J Alzheimers Dis. 2020;78(2):683–97.PubMedCrossRef Marizzoni M, et al. Short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer’s disease. J Alzheimers Dis. 2020;78(2):683–97.PubMedCrossRef
108.
go back to reference Hu L, et al. High salt elicits brain inflammation and cognitive dysfunction, accompanied by alternations in the gut microbiota and decreased SCFA production. J Alzheimers Dis. 2020;77(2):629–40.PubMedCrossRef Hu L, et al. High salt elicits brain inflammation and cognitive dysfunction, accompanied by alternations in the gut microbiota and decreased SCFA production. J Alzheimers Dis. 2020;77(2):629–40.PubMedCrossRef
109.
go back to reference Goyal D, Ali SA, Singh RK. Emerging role of gut microbiota in modulation of neuroinflammation and neurodegeneration with emphasis on Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2021;106:110112.PubMedCrossRef Goyal D, Ali SA, Singh RK. Emerging role of gut microbiota in modulation of neuroinflammation and neurodegeneration with emphasis on Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2021;106:110112.PubMedCrossRef
110.
go back to reference Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motility. 2019;25(1):48.CrossRef Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motility. 2019;25(1):48.CrossRef
112.
go back to reference Cuervo-Zanatta D, Garcia-Mena J, Perez-Cruz C. Gut microbiota alterations and cognitive impairment are sexually dissociated in a transgenic mice model of Alzheimer’s disease. J Alzheimers Dis. 2021;82(s1):S195-s214.PubMedCrossRef Cuervo-Zanatta D, Garcia-Mena J, Perez-Cruz C. Gut microbiota alterations and cognitive impairment are sexually dissociated in a transgenic mice model of Alzheimer’s disease. J Alzheimers Dis. 2021;82(s1):S195-s214.PubMedCrossRef
113.
114.
115.
go back to reference Arora K, Green M, Prakash S. The microbiome and Alzheimer’s disease: potential and limitations of prebiotic, synbiotic, and probiotic formulations. Front Bioengineering Biotechnol. 2020;8:1411.CrossRef Arora K, Green M, Prakash S. The microbiome and Alzheimer’s disease: potential and limitations of prebiotic, synbiotic, and probiotic formulations. Front Bioengineering Biotechnol. 2020;8:1411.CrossRef
116.
go back to reference Zhan X, Stamova B, Sharp FR. Lipopolysaccharide associates with amyloid plaques, neurons and oligodendrocytes in alzheimer’s disease brain: a review. Front Aging Neurosci. 2018;10:42–42.PubMedPubMedCentralCrossRef Zhan X, Stamova B, Sharp FR. Lipopolysaccharide associates with amyloid plaques, neurons and oligodendrocytes in alzheimer’s disease brain: a review. Front Aging Neurosci. 2018;10:42–42.PubMedPubMedCentralCrossRef
117.
go back to reference Zhang J-C, Yao W, Hashimoto K. Brain-derived neurotrophic factor (BDNF)-TrkB signaling in inflammation-related depression and potential therapeutic targets. Curr Neuropharmacol. 2016;14(7):721–31.PubMedPubMedCentralCrossRef Zhang J-C, Yao W, Hashimoto K. Brain-derived neurotrophic factor (BDNF)-TrkB signaling in inflammation-related depression and potential therapeutic targets. Curr Neuropharmacol. 2016;14(7):721–31.PubMedPubMedCentralCrossRef
118.
119.
go back to reference Zhou J, et al. Imbalance of microglial TLR4/TREM2 in LPS-Treated APP/PS1 transgenic mice: a potential link between Alzheimer’s disease and systemic inflammation. Neurochem Res. 2019;44(5):1138–51.PubMedCrossRef Zhou J, et al. Imbalance of microglial TLR4/TREM2 in LPS-Treated APP/PS1 transgenic mice: a potential link between Alzheimer’s disease and systemic inflammation. Neurochem Res. 2019;44(5):1138–51.PubMedCrossRef
120.
go back to reference Ghosh S, et al. Sustained interleukin-1β overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33(11):5053–64.PubMedPubMedCentralCrossRef Ghosh S, et al. Sustained interleukin-1β overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33(11):5053–64.PubMedPubMedCentralCrossRef
121.
go back to reference Garcez ML, Jacobs KR, Guillemin GJ. Microbiota alterations in Alzheimer’s disease: involvement of the kynurenine pathway and inflammation. Neurotox Res. 2019;36(2):424–36.PubMedCrossRef Garcez ML, Jacobs KR, Guillemin GJ. Microbiota alterations in Alzheimer’s disease: involvement of the kynurenine pathway and inflammation. Neurotox Res. 2019;36(2):424–36.PubMedCrossRef
123.
go back to reference Hort J, Valis M, Angelucci F. Administration of pre/probiotics with conventional drug treatment in Alzheimer’s disease. Neural Regen Res. 2020;15(3):448.PubMedCrossRef Hort J, Valis M, Angelucci F. Administration of pre/probiotics with conventional drug treatment in Alzheimer’s disease. Neural Regen Res. 2020;15(3):448.PubMedCrossRef
124.
go back to reference Tamtaji OR, et al. Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in Alzheimer’s disease: a randomized, double-blind, controlled trial. Clin Nutr. 2019;38(6):2569–75.PubMedCrossRef Tamtaji OR, et al. Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in Alzheimer’s disease: a randomized, double-blind, controlled trial. Clin Nutr. 2019;38(6):2569–75.PubMedCrossRef
125.
go back to reference Buford TW, et al. Angiotensin (1–7) delivered orally via probiotic, but not subcutaneously, benefits the gut-brain axis in older rats. Geroscience. 2020;42(5):1307–21.PubMedPubMedCentralCrossRef Buford TW, et al. Angiotensin (1–7) delivered orally via probiotic, but not subcutaneously, benefits the gut-brain axis in older rats. Geroscience. 2020;42(5):1307–21.PubMedPubMedCentralCrossRef
126.
go back to reference Carter CS, et al. Therapeutic delivery of ang(1–7) via genetically modified probiotic: a dosing study. J Gerontol A Biol Sci Med Sci. 2020;75(7):1299–303.PubMedCrossRef Carter CS, et al. Therapeutic delivery of ang(1–7) via genetically modified probiotic: a dosing study. J Gerontol A Biol Sci Med Sci. 2020;75(7):1299–303.PubMedCrossRef
127.
go back to reference Machado AS, et al. Oral probiotic bifidobacterium longum supplementation improves metabolic parameters and alters the expression of the renin-angiotensin system in obese mice liver. Biol Res Nurs. 2021;23(1):100–8.PubMedCrossRef Machado AS, et al. Oral probiotic bifidobacterium longum supplementation improves metabolic parameters and alters the expression of the renin-angiotensin system in obese mice liver. Biol Res Nurs. 2021;23(1):100–8.PubMedCrossRef
128.
129.
130.
go back to reference Armstrong MJ, Okun MS. Diagnosis and treatment of parkinson disease: a review. JAMA. 2020;323(6):548–60.PubMedCrossRef Armstrong MJ, Okun MS. Diagnosis and treatment of parkinson disease: a review. JAMA. 2020;323(6):548–60.PubMedCrossRef
131.
go back to reference Jankovic J, Tan EK. Parkinson’s disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020;91(8):795–808.PubMedCrossRef Jankovic J, Tan EK. Parkinson’s disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020;91(8):795–808.PubMedCrossRef
132.
go back to reference Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics, and pathophysiology. Clin Geriatr Med. 2020;36(1):1–12.PubMedCrossRef Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics, and pathophysiology. Clin Geriatr Med. 2020;36(1):1–12.PubMedCrossRef
133.
go back to reference Mertens B, et al. The role of the central renin-angiotensin system in Parkinson’s disease. J Renin Angiotensin Aldosterone Syst. 2010;11(1):49–56.PubMedCrossRef Mertens B, et al. The role of the central renin-angiotensin system in Parkinson’s disease. J Renin Angiotensin Aldosterone Syst. 2010;11(1):49–56.PubMedCrossRef
134.
go back to reference Kobiec T, et al. The renin-angiotensin system modulates dopaminergic neurotransmission: a new player on the scene. Front Synaptic Neurosci. 2021;13:638519.PubMedPubMedCentralCrossRef Kobiec T, et al. The renin-angiotensin system modulates dopaminergic neurotransmission: a new player on the scene. Front Synaptic Neurosci. 2021;13:638519.PubMedPubMedCentralCrossRef
135.
go back to reference Rodriguez-Pallares J, et al. Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS. Neurobiol Dis. 2008;31(1):58–73.PubMedCrossRef Rodriguez-Pallares J, et al. Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS. Neurobiol Dis. 2008;31(1):58–73.PubMedCrossRef
136.
137.
go back to reference Labandeira-García JL, et al. Brain renin-angiotensin system and dopaminergic cell vulnerability. Front Neuroanat. 2014;8:67.PubMedPubMedCentral Labandeira-García JL, et al. Brain renin-angiotensin system and dopaminergic cell vulnerability. Front Neuroanat. 2014;8:67.PubMedPubMedCentral
138.
go back to reference Nishiwaki H, et al. Meta-analysis of gut dysbiosis in Parkinson’s Disease. Mov Disord. 2020;35(9):1626–35.PubMedCrossRef Nishiwaki H, et al. Meta-analysis of gut dysbiosis in Parkinson’s Disease. Mov Disord. 2020;35(9):1626–35.PubMedCrossRef
140.
go back to reference Cirstea MS, et al. Microbiota composition and metabolism are associated with gut function in Parkinson’s disease. Mov Disord. 2020;35(7):1208–17.PubMedCrossRef Cirstea MS, et al. Microbiota composition and metabolism are associated with gut function in Parkinson’s disease. Mov Disord. 2020;35(7):1208–17.PubMedCrossRef
141.
go back to reference Petrov VA, et al. Analysis of gut microbiota in patients with Parkinson’s disease. Bull Exp Biol Med. 2017;162(6):734–7.PubMedCrossRef Petrov VA, et al. Analysis of gut microbiota in patients with Parkinson’s disease. Bull Exp Biol Med. 2017;162(6):734–7.PubMedCrossRef
142.
143.
go back to reference Qian Y, et al. Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav Immun. 2018;70:194–202.PubMedCrossRef Qian Y, et al. Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav Immun. 2018;70:194–202.PubMedCrossRef
144.
go back to reference Takahashi K, et al. Altered gut microbiota in Parkinson’s disease patients with motor complications. Parkinsonism Relat Disord. 2021;95:11–7.PubMedCrossRef Takahashi K, et al. Altered gut microbiota in Parkinson’s disease patients with motor complications. Parkinsonism Relat Disord. 2021;95:11–7.PubMedCrossRef
145.
go back to reference Yan Z, et al. Alterations of gut microbiota and metabolome with Parkinson’s disease. Microb Pathog. 2021;160:105187.PubMedCrossRef Yan Z, et al. Alterations of gut microbiota and metabolome with Parkinson’s disease. Microb Pathog. 2021;160:105187.PubMedCrossRef
146.
go back to reference Unger MM, et al. 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.PubMedCrossRef Unger MM, et al. 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.PubMedCrossRef
147.
go back to reference Huang T, et al. The gut microbiota metabolite propionate ameliorates intestinal epithelial barrier dysfunction-mediated Parkinson’s disease via the AKT signaling pathway. NeuroReport. 2021;32(3):244–51.PubMedCrossRef Huang T, et al. The gut microbiota metabolite propionate ameliorates intestinal epithelial barrier dysfunction-mediated Parkinson’s disease via the AKT signaling pathway. NeuroReport. 2021;32(3):244–51.PubMedCrossRef
149.
go back to reference Sharma S, Awasthi A, Singh S. Altered gut microbiota and intestinal permeability in Parkinson’s disease: pathological highlight to management. Neurosci Lett. 2019;712:134516.PubMedCrossRef Sharma S, Awasthi A, Singh S. Altered gut microbiota and intestinal permeability in Parkinson’s disease: pathological highlight to management. Neurosci Lett. 2019;712:134516.PubMedCrossRef
150.
go back to reference Dutta SK, et al. Parkinson’s disease: the emerging role of gut dysbiosis, antibiotics, probiotics, and fecal microbiota transplantation. J Neurogastroenterol Motility. 2019;25(3):363.CrossRef Dutta SK, et al. Parkinson’s disease: the emerging role of gut dysbiosis, antibiotics, probiotics, and fecal microbiota transplantation. J Neurogastroenterol Motility. 2019;25(3):363.CrossRef
153.
go back to reference Chen Z-J, et al. Association of Parkinson’s disease with microbes and microbiological therapy. Front Cell Infect Microbiol. 2021;11:93. Chen Z-J, et al. Association of Parkinson’s disease with microbes and microbiological therapy. Front Cell Infect Microbiol. 2021;11:93.
155.
go back to reference Sun MF, et al. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav Immun. 2018;70:48–60.PubMedCrossRef Sun MF, et al. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav Immun. 2018;70:48–60.PubMedCrossRef
156.
go back to reference Castelli V, et al. The emerging role of probiotics in neurodegenerative diseases: new hope for Parkinson’s disease? Neural Regen Res. 2021;16(4):628.PubMedCrossRef Castelli V, et al. The emerging role of probiotics in neurodegenerative diseases: new hope for Parkinson’s disease? Neural Regen Res. 2021;16(4):628.PubMedCrossRef
157.
go back to reference Severance EG, et al. Probiotic normalization of Candida albicans in schizophrenia: a randomized, placebo-controlled, longitudinal pilot study. Brain Behav Immun. 2017;62:41–5.PubMedCrossRef Severance EG, et al. Probiotic normalization of Candida albicans in schizophrenia: a randomized, placebo-controlled, longitudinal pilot study. Brain Behav Immun. 2017;62:41–5.PubMedCrossRef
158.
go back to reference Rudzki L, et al. 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;100:213–22.PubMedCrossRef Rudzki L, et al. 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;100:213–22.PubMedCrossRef
159.
go back to reference Tian P, et al. Towards a psychobiotic therapy for depression: Bifidobacterium breve CCFM1025 reverses chronic stress-induced depressive symptoms and gut microbial abnormalities in mice. Neurobiol Stress. 2020;12:100216.PubMedPubMedCentralCrossRef Tian P, et al. Towards a psychobiotic therapy for depression: Bifidobacterium breve CCFM1025 reverses chronic stress-induced depressive symptoms and gut microbial abnormalities in mice. Neurobiol Stress. 2020;12:100216.PubMedPubMedCentralCrossRef
160.
go back to reference Chen H-M, et al. Psychophysiological effects of lactobacillus plantarum ps128 in patients with major depressive disorder: a preliminary 8-week open trial. Nutrients. 2021;13(11):3731.PubMedPubMedCentralCrossRef Chen H-M, et al. Psychophysiological effects of lactobacillus plantarum ps128 in patients with major depressive disorder: a preliminary 8-week open trial. Nutrients. 2021;13(11):3731.PubMedPubMedCentralCrossRef
161.
go back to reference Dickerson F, et al. Adjunctive probiotic microorganisms to prevent rehospitalization in patients with acute mania: a randomized controlled trial. Bipolar Disord. 2018;20(7):614–21.PubMedCrossRef Dickerson F, et al. Adjunctive probiotic microorganisms to prevent rehospitalization in patients with acute mania: a randomized controlled trial. Bipolar Disord. 2018;20(7):614–21.PubMedCrossRef
162.
go back to reference Kim C-S, et al. 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 Series A. 2021;76(1):32–40.CrossRef Kim C-S, et al. 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 Series A. 2021;76(1):32–40.CrossRef
163.
go back to reference Cussotto S, et al. The neuroendocrinology of the microbiota-gut-brain axis: a behavioural perspective. Front Neuroendocrinol. 2018;51:80–101.PubMedCrossRef Cussotto S, et al. The neuroendocrinology of the microbiota-gut-brain axis: a behavioural perspective. Front Neuroendocrinol. 2018;51:80–101.PubMedCrossRef
164.
go back to reference Fülling C, Dinan TG, Cryan JF. Gut microbe to brain signaling: what happens in vagus. Neuron. 2019;101(6):998–1002.PubMedCrossRef Fülling C, Dinan TG, Cryan JF. Gut microbe to brain signaling: what happens in vagus. Neuron. 2019;101(6):998–1002.PubMedCrossRef
165.
go back to reference Jaggar M, et al. You’ve got male: sex and the microbiota-gut-brain axis across the lifespan. Front Neuroendocrinol. 2020;56:100815.PubMedCrossRef Jaggar M, et al. You’ve got male: sex and the microbiota-gut-brain axis across the lifespan. Front Neuroendocrinol. 2020;56:100815.PubMedCrossRef
166.
go back to reference Rouanet A, et al. Live biotherapeutic products, a road map for safety assessment. Front Med. 2020;7:237.CrossRef Rouanet A, et al. Live biotherapeutic products, a road map for safety assessment. Front Med. 2020;7:237.CrossRef
Metadata
Title
Recent developments in the probiotics as live biotherapeutic products (LBPs) as modulators of gut brain axis related neurological conditions
Authors
Duygu Ağagündüz
Feray Gençer Bingöl
Elif Çelik
Özge Cemali
Çiler Özenir
Fatih Özoğul
Raffaele Capasso
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2022
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03609-y

Other articles of this Issue 1/2022

Journal of Translational Medicine 1/2022 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.