Prebiotic and Probiotic Modulation of the Microbiota–Gut–Brain Axis in Depression
Abstract
:1. Introduction
2. The Gut Microbiome
3. The Microbiota–Gut–Brain Axis: Putative Mechanisms of Bug-to-Brain Communications
3.1. The Microbiota–Gut–Brain Axis Communicates through the Vagus Nerve
3.2. The Microbiota–Gut–Brain Axis Communicates through the Immune System
3.3. Microbiota–Gut–Brain Axis Communicates through Circulating Gut-Derived Microbial Metabolites and Neurotransmitters
4. Perturbed Microbiota–Gut–Brain Axis Metabolism in Mood Disorders
4.1. Profiling Gut Microbiota Changes in Mood Disorders: The Way Forward
4.2. Animal Studies
4.3. Human Studies
5. Modulation of the Microbiota–Gut–Brain Axis by Prebiotics and Probiotics
5.1. Prebiotics Modulate Brain Function and Behaviour in Animal Models
5.2. Probiotics Modulate Brain Function and Behaviour in Animal Models
Model Organism (Paradigm) | Probiotic (Dose) | Duration of Treatment | Behavioural Effects | Neurochemical Effects | Reference |
---|---|---|---|---|---|
7-week-old male BALB/c mice (healthy) | B. longum 1714 or B. breve 1205 (1 × 109 CFU, daily) | 3 weeks prior to start of behavioural tests, 3 weeks during behavioural testing (6 weeks total) | ↓ anxiety (OFT, MBT) and ↓ depressive-like behaviour (FST) (B. longum 1714) ↓ anxiety (EPM and MBT) (B. breve 1205) | Not applicable | [95] |
6-week-old male BALB/c mice (vagotomy) | L. rhamnosus JB-1 (1 × 109 CFU, daily) | 4 weeks (behavioural assays started during final week of treatment) | ↓ anxiety (EPM, OFT) ↓ depressive-like behaviour (FST) | Altered hippocampal GABAA receptor subunit expression, effects dependent on vagus nerve | [35] |
4-week-old male BALB/c mice (healthy MRI and MRS brain imaging) | L. rhamnosus JB-1 (1 × 109 CFU, daily) | 4 weeks (imaging performed throughout duration of treatment at weeks 0, 1, 2, 3, 4, and 4 weeks post-treatment cessation) | Not applicable | ↑ brain Glx at weeks 2, 3, 4, 8 ↑ brain NAA at weeks 1, 2, 3, 4 ↑ brain GABA at week 4 only | [96] |
Male Swiss albino mice 25–30 g (28 days CMS) | L. plantarum MTCC 9510 (2 × 1010 CFU, daily) | 4 weeks (duration of CMS paradigm) | CMS ↓ locomotion (OFT), ↑ depressive-like behaviour (FST, TST), reversed by L. plantarum MTCC 9510 | CMS ↑ BBB permeability, ↑ brain NF-kB, reversed by L. plantarum MTCC 9510 | [97] |
Male neonatal C57Bl/6 mice (maternal separation) | B. pseudocatenulatum CECT 7765 (1 × 108 CFU, daily) | 20 days (PND2 to PND21) | MS ↑ anxiety (EPM). B. pseudocatenulatum CECT 7765 ↓ anxiety (EPM) | Not applicable | [98] |
4-week-old male Sprague Dawley rats (chronic HFD, 60% of kilocalories from fat) | B. bifidum W23, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lc. Lactis W19, Lc. Lactis W58 (3.75 × 108 CFU/mL in drinking water, daily) | 5 weeks prior to start of behavioural tests, 10 weeks total | Probiotic mixture ↓ depressive-like behaviour (FST) irrespective of diet. | Not applicable | [99] |
5.3. Prebiotics and Probiotics in Human Studies
Paradigm | Probiotic (Dose) | Duration of Treatment | Effects | Reference |
---|---|---|---|---|
Adult males and females (healthy) | L. helveticus R0052 and B. longum R0175 (3 × 109 CFU, daily) | 30 days | ↓ anxiety and depression HAD scores No effect on BDI or BAI scores | [111] |
Adult males (healthy) | L. rhamnosus JB-1 (1 × 109 CFU, daily) | 4 weeks | No differences between probiotic and placebo for BDI, BDAI, or serum cytokine levels | [112] |
Adult males and females (mild to moderate depression: PHQ9 score 5–19) | Bacillus subtilis, Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium infantis, Bifidobacterium longum, Lactobacillus acidophilus, Lactobacillus delbrueckii ssp. Bulgaricus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus helveticus, Lactobacillus salivarius, Lactococcus lactis ssp. Lactis, Streptococcus thermophilus. (2 × 109 CFU, daily) | 4 weeks | No effect of probiotic on salivary cortisol or serum CRP Probiotic ↓ PHQ9 scores | [113] |
Adult males and females (moderate or severe depression: ≥11 QIDS-SR16 or ≥14 DASS-42) | L. helveticus R0052 and B. longum R0175 (3 × 109 CFU, daily) | 8 weeks | No effects of probiotic treatment on self-reported depression or anxiety scales, and no effect on serum CRP, IL-1B, IL-6, TNF, BDNF, or vitamin D | [114] |
Adult males and females (MDD diagnosis, moderate-severe symptoms) | L. acidophilus, L. casei, and B. bifidum (6 × 109 CFU, daily) | 8 weeks | Probiotic ↓ BDI score compared to placebo Probiotic ↓ serum CRP level compared to placebo | [115] |
Adult males and females (MDD diagnosis, mild-moderate symptoms) | L. helveticus R0052 and B. longum R0175 (1 × 1010 CFU, daily) | 8 weeks | Probiotic ↓ BDI score compared to placebo Probiotic ↓ serum kynurenine/tryptophan ratio compared to placebo | [109] |
Adult pregnant women (at least mild anxiety or depression: ≥10 EPDS or ≥40 STAI) | B. bifidum W23, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lc. Lactis W19, Lc. Lactis W58 (5 × 109 CFU, daily) | From 26 to 30 weeks gestation until delivery | No effect of probiotic on perinatal maternal depression (EPDS) or anxiety (STAI) scores | [116] |
Adult pregnant women (healthy) | L. rhamnosus HN001 (6 × 109 CFU, daily) | From 14 to 16 weeks gestation until 6 months postpartum | Probiotic significantly ↓ depression (EPDS) and anxiety (STAI) scores compared to placebo | [117] |
Adult pregnant women (obese: BMI ≥ 30 kg/m2) | L. rhamnosus GG and B. lactis BB12 (6.5 × 109 CFU, daily) | From 12 to 18 weeks gestation until 36 weeks gestation | No effect of probiotic on depression (EPDS) or anxiety (STAI-6) scores compared to placebo | [118] |
5.4. Prebiotics and Probiotics during the Perinatal Period
6. Gastrointestinal Engraftment of Probiotic Supplements
7. Depression and Obesity: A Bidirectional Relationship
8. Conclusion and Perspectives for Future Research
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Conflicts of Interest
References
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W. Psychobiotics and the Manipulation of Bacteria–Gut–Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
- Malhi, G.S.; Mann, J.J. Depression. Lancet 2018, 392, 2299–2312. [Google Scholar] [CrossRef] [PubMed]
- Jorm, A.F.; Patten, S.B.; Brugha, T.S.; Mojtabai, R. Has increased provision of treatment reduced the prevalence of common mental disorders? Review of the evidence from four countries. World Psychiatry 2017, 16, 90–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ilyas, S.; Moncrieff, J. Trends in prescriptions and costs of drugs for mental disorders in England, 1998–2010. Br. J. Psychiatry 2012, 200, 393–398. [Google Scholar] [CrossRef] [PubMed]
- Iacobucci, G. NHS prescribed record number of antidepressants last year. BMJ 2019, 364, l1508. [Google Scholar] [CrossRef] [PubMed]
- Baker, C. Mental Health Statistics: Prevalence, Services and Funding in England. 2021. Available online: https://commonslibrary.parliament.uk/research-briefings/sn06988/ (accessed on 28 April 2022).
- 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] [Green Version]
- Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef]
- Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2015, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
- After the Integrative Human Microbiome Project, what’s next for the microbiome community? Nature 2019, 569, 599. [CrossRef] [Green Version]
- Lloyd-Price, J.; Mahurkar, A.; Rahnavard, G.; Crabtree, J.; Orvis, J.; Hall, A.B.; Brady, A.; Creasy, H.H.; McCracken, C.; Giglio, M.G.; et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature 2017, 550, 61–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
- Fettweis, J.M.; Serrano, M.G.; Brooks, J.P.; Edwards, D.J.; Girerd, P.H.; Parikh, H.I.; Huang, B.; Arodz, T.J.; Edupuganti, L.; Glascock, A.L.; et al. The vaginal microbiome and preterm birth. Nat. Med. 2019, 25, 1012–1021. [Google Scholar] [CrossRef] [Green Version]
- Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef] [PubMed]
- Zhou, W.; Sailani, M.R.; Contrepois, K.; Zhou, Y.; Ahadi, S.; Leopold, S.R.; Zhang, M.J.; Rao, V.; Avina, M.; Mishra, T.; et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature 2019, 569, 663–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gaulke, C.A.; Sharpton, T.J. The influence of ethnicity and geography on human gut microbiome composition. Nat. Med. 2018, 24, 1495–1496. [Google Scholar] [CrossRef]
- Finding diversity in the microbiome. Nat. Med. 2019, 25, 863. [CrossRef] [Green Version]
- Hegstrand, L.R.; Hine, R.J. Variations of brain histamine levels in germ-free and nephrectomized rats. Neurochem. Res. 1986, 11, 185–191. [Google Scholar] [CrossRef]
- Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.-N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef]
- Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [Green Version]
- Gareau, M.G.; Wine, E.; Rodrigues, D.M.; Cho, J.H.; Whary, M.T.; Philpott, D.J.; MacQueen, G.M.; Sherman, P.M. Bacterial infection causes stress-induced memory dysfunction in mice. Gut 2011, 60, 307–317. [Google Scholar] [CrossRef]
- Neufeld, K.M.; Kang, N.; Bienenstock, J.; Foster, J.A. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol. Motil. 2011, 23, 255-e119. [Google Scholar] [CrossRef]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
- Forsythe, P.; Bienenstock, J.; Kunze, W.A. Vagal pathways for microbiome-brain-gut axis communication. Adv. Exp. Med. Biol. 2014, 817, 115–133. [Google Scholar] [CrossRef]
- Sinclair-Gieben, A.H.C.; Clark, C.G.; Dean, A.C.B. Psychiatric Illness following Surgery for Duodenal Ulcer. Scott. Med. J. 1962, 7, 168–172. [Google Scholar] [CrossRef] [PubMed]
- Kral, J. Vagotomy for treatment of severe obesity. Lancet 1978, 311, 307–308. [Google Scholar] [CrossRef]
- Kral, J.G. Behavioral effects of vagotomy in humans. J. Auton. Nerv. Syst. 1983, 9, 273–281. [Google Scholar] [CrossRef] [PubMed]
- Aaronson, S.T.; Sears, P.; Ruvuna, F.; Bunker, M.; Conway, C.R.; Dougherty, D.D.; Reimherr, F.W.; Schwartz, T.L.; Zajecka, J.M. A 5-Year Observational Study of Patients with Treatment-Resistant Depression Treated with Vagus Nerve Stimulation or Treatment as Usual: Comparison of Response, Remission, and Suicidality. Am. J. Psychiatry 2017, 174, 640–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Layé, S.; Bluthé, R.M.; Kent, S.; Combe, C.; Medina, C.; Parnet, P.; Kelley, K.; Dantzer, R. Subdiaphragmatic vagotomy blocks induction of IL-1 beta mRNA in mice brain in response to peripheral LPS. Am. J. Physiol. Integr. Comp. Physiol. 1995, 268, R1327–R1331. [Google Scholar] [CrossRef]
- Bluthé, R.M.; Walter, V.; Parnet, P.; Layé, S.; Lestage, J.; Verrier, D.; Poole, S.; Stenning, B.E.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide induces sickness behaviour in rats by a vagal mediated mechanism. Comptes Rendus Acad. Sci. III 1994, 317, 499–503. [Google Scholar]
- Bretdibat, J.; Bluthe, R.; Kent, S.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide and Interleukin-1 depress food-motivated behavior in mice by a vagal-mediated mechanism. Brain Behav. Immun. 1995, 9, 242–246. [Google Scholar] [CrossRef] [Green Version]
- Van Dam, A.-M.; Bol, J.G.; Gaykema, R.P.; Goehler, L.E.; Maier, S.F.; Watkins, L.R.; Tilders, F.J. Vagotomy does not inhibit high dose lipopolysaccharide-induced interleukin-1β immunoreactivity in rat brain and pituitary gland. Neurosci. Lett. 2000, 285, 169–172. [Google Scholar] [CrossRef] [PubMed]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bercik, P.; Park, A.J.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; Deng, Y.; Blennerhassett, P.A.; Fahnestock, M.; Moine, D.; et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol. Motil. 2011, 23, 1132–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pitharouli, M.C.; Hagenaars, S.P.; Glanville, K.P.; Coleman, J.R.; Hotopf, M.; Lewis, C.M.; Pariante, C.M. Elevated C-Reactive Protein in Patients with Depression, Independent of Genetic, Health, and Psychosocial Factors: Results from the UK Biobank. Am. J. Psychiatry 2021, 178, 522–529. [Google Scholar] [CrossRef]
- Boeschoten, R.E.; Braamse, A.M.J.; Beekman, A.T.F.; Cuijpers, P.; van Oppen, P.; Dekker, J.; Uitdehaag, B.M. Prevalence of depression and anxiety in Multiple Sclerosis: A systematic review and meta-analysis. J. Neurol. Sci. 2017, 372, 331–341. [Google Scholar] [CrossRef]
- Keefer, L.; Kane, S.V. Considering the Bidirectional Pathways Between Depression and IBD: Recommendations for Comprehensive IBD Care. Gastroenterol. Hepatol. 2017, 13, 164–169. [Google Scholar]
- Labenz, C.; Huber, Y.; Michel, M.; Nagel, M.; Galle, P.R.; Kostev, K.; Schattenberg, J.M. Nonalcoholic Fatty Liver Disease Increases the Risk of Anxiety and Depression. Hepatol. Commun. 2020, 4, 1293–1301. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Powell, N.; Walker, M.M.; Talley, N.J. The mucosal immune system: Master regulator of bidirectional gut–brain communications. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 143–159. [Google Scholar] [CrossRef] [PubMed]
- Shan, M.; Gentile, M.; Yeiser, J.R.; Walland, A.C.; Bornstein, V.U.; Chen, K.; He, B.; Cassis, L.; Bigas, A.; Cols, M.; et al. Mucus enhances gut homeostasis and oral tolerance by delivering immunoregulatory signals. Science 2013, 342, 447–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Macpherson, A.J.; Uhr, T. Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science 2004, 303, 1662–1665. [Google Scholar] [CrossRef] [Green Version]
- Peterson, D.A.; McNulty, N.P.; Guruge, J.L.; Gordon, J.I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2007, 2, 328–339. [Google Scholar] [CrossRef] [Green Version]
- Faria, A.M.C.; Weiner, H.L. Oral tolerance. Immunol. Rev. 2005, 206, 232–259. [Google Scholar] [CrossRef]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed]
- Mossad, O.; Batut, B.; Yilmaz, B.; Dokalis, N.; Mezö, C.; Nent, E.; Nabavi, L.S.; Mayer, M.; Maron, F.J.M.; Buescher, J.M.; et al. Gut microbiota drives age-related oxidative stress and mitochondrial damage in microglia via the metabolite N6-carboxymethyllysine. Nat. Neurosci. 2022, 25, 295–305. [Google Scholar] [CrossRef] [PubMed]
- Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698–3703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, Y.; Liu, C.-W.; Yang, Y.; Hsiao, Y.-C.; Ru, H.; Lu, K. High-coverage metabolomics uncovers microbiota-driven biochemical landscape of interorgan transport and gut-brain communication in mice. Nat. Commun. 2021, 12, 6000. [Google Scholar] [CrossRef]
- Chu, C.; Murdock, M.H.; Jing, D.; Won, T.H.; Chung, H.; Kressel, A.M.; Tsaava, T.; Addorisio, M.E.; Putzel, G.G.; Zhou, L.; et al. The microbiota regulate neuronal function and fear extinction learning. Nature 2019, 574, 543–548. [Google Scholar] [CrossRef]
- Vuong, H.E.; Pronovost, G.N.; Williams, D.W.; Coley, E.J.L.; Siegler, E.L.; Qiu, A.; Kazantsev, M.; Wilson, C.J.; Rendon, T.; Hsiao, E.Y. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020, 586, 281–286. [Google Scholar] [CrossRef]
- Valle-Gough, R.E.; Samaniego-Gámez, B.Y.; Apodaca-Hernández, J.E.; Chiappa-Carrara, F.X.; Rodríguez-Dorantes, M.; Arena-Ortiz, M.L. RNA-Seq Analysis on the Microbiota Associated with the White Shrimp (Litopenaeus vannamei) in Different Stages of Development. Appl. Sci. 2022, 12, 2483. [Google Scholar] [CrossRef]
- Buffet-Bataillon, S.; Rizk, G.; Cattoir, V.; Sassi, M.; Thibault, V.; Del Giudice, J.; Gangneux, J.-P. Efficient and Quality-Optimized Metagenomic Pipeline Designed for Taxonomic Classification in Routine Microbiological Clinical Tests. Microorganisms 2022, 10, 711. [Google Scholar] [CrossRef] [PubMed]
- Karaduta, O.; Dvanajscak, Z.; Zybailov, B. Metaproteomics—An Advantageous Option in Studies of Host-Microbiota Interaction. Microorganisms 2021, 9, 980. [Google Scholar] [CrossRef]
- Cheung, S.G.; Goldenthal, A.R.; Uhlemann, A.-C.; Mann, J.J.; Miller, J.M.; Sublette, M.E. Systematic Review of Gut Microbiota and Major Depression. Front. Psychiatry 2019, 10, 34. [Google Scholar] [CrossRef] [Green Version]
- Nikolova, V.L.; Smith, M.R.B.; Hall, L.J.; Cleare, A.J.; Stone, J.M.; Young, A.H. Perturbations in Gut Microbiota Composition in Psychiatric Disorders. JAMA Psychiatry 2021, 78, 1343. [Google Scholar] [CrossRef] [PubMed]
- Zierer, J.; Jackson, M.A.; Kastenmüller, G.; Mangino, M.; Long, T.; Telenti, A.; Mohney, R.P.; Small, K.S.; Bell, J.T.; Steves, C.J.; et al. The fecal metabolome as a functional readout of the gut microbiome. Nat. Genet. 2018, 50, 790–795. [Google Scholar] [CrossRef] [PubMed]
- Visconti, A.; Le Roy, C.I.; Rosa, F.; Rossi, N.; Martin, T.C.; Mohney, R.P.; Li, W.; de Rinaldis, E.; Bell, J.T.; Venter, J.C.; et al. Interplay between the human gut microbiome and host metabolism. Nat. Commun. 2019, 10, 4505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed] [Green Version]
- Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.-J.; Fan, S.-H.; Du, X.; et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef] [PubMed]
- Cummings, K.A.; Popescu, G.K. Glycine-dependent activation of NMDA receptors. J. Gen. Physiol. 2015, 145, 513–527. [Google Scholar] [CrossRef] [Green Version]
- Labrie, V.; Roder, J.C. The involvement of the NMDA receptor d-serine/glycine site in the pathophysiology and treatment of schizophrenia. Neurosci. Biobehav. Rev. 2010, 34, 351–372. [Google Scholar] [CrossRef] [PubMed]
- Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109–118. [Google Scholar] [CrossRef]
- Pu, J.; Liu, Y.; Gui, S.; Tian, L.; Yu, Y.; Song, X.; Zhong, X.; Chen, X.; Chen, W.; Zheng, P.; et al. Metabolomic changes in animal models of depression: A systematic analysis. Mol. Psychiatry 2021, 26, 7328–7336. [Google Scholar] [CrossRef]
- Pu, J.; Yu, Y.; Liu, Y.; Tian, L.; Gui, S.; Zhong, X.; Fan, C.; Xu, S.; Song, X.; Liu, L.; et al. MENDA: A comprehensive curated resource of metabolic characterization in depression. Briefings Bioinform. 2020, 21, 1455–1464. [Google Scholar] [CrossRef] [Green Version]
- López-López, A.L.; Jaime, H.B.; Villanueva, M.D.C.E.; Padilla, M.B.; Palacios, G.V.; Aguilar, F.J.A. Chronic unpredictable mild stress generates oxidative stress and systemic inflammation in rats. Physiol. Behav. 2016, 161, 15–23. [Google Scholar] [CrossRef]
- Pavlov, D.; Bettendorff, L.; Gorlova, A.; Olkhovik, A.; Kalueff, A.V.; Ponomarev, E.D.; Inozemtsev, A.; Chekhonin, V.; Lesch, K.-P.; Anthony, D.C.; et al. Neuroinflammation and aberrant hippocampal plasticity in a mouse model of emotional stress evoked by exposure to ultrasound of alternating frequencies. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 90, 104–116. [Google Scholar] [CrossRef] [PubMed]
- Kaufman, J.; Charney, D. Comorbidity of mood and anxiety disorders. Depress. Anxiety 2000, 12, 69–76. [Google Scholar] [CrossRef] [PubMed]
- Needham, B.D.; Funabashi, M.; Adame, M.D.; Wang, Z.; Boktor, J.C.; Haney, J.; Wu, W.-L.; Rabut, C.; Ladinsky, M.S.; Hwang, S.-J.; et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647–653. [Google Scholar] [CrossRef] [PubMed]
- Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linløkken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 1155–1162. [Google Scholar] [CrossRef]
- Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saulnier, D.M.; Riehle, K.; Mistretta, T.A.; Diaz, M.A.; Mandal, D.; Raza, S.; Weidler, E.M.; Qin, X.; Coarfa, C.; Milosavljevic, A.; et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology 2011, 141, 1782–1791. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Zheng, P.; Li, Y.; Wu, J.; Tan, X.; Zhou, J.; Sun, Z.; Chen, X.; Zhang, G.; Zhang, H.; et al. Landscapes of bacterial and metabolic signatures and their interaction in major depressive disorders. Sci. Adv. 2020, 6, eaba8555. [Google Scholar] [CrossRef]
- Li, Z.; Lai, J.; Zhang, P.; Ding, J.; Jiang, J.; Liu, C.; Huang, H.; Zhen, H.; Xi, C.; Sun, Y.; et al. Multi-omics analyses of serum metabolome, gut microbiome and brain function reveal dysregulated microbiota-gut-brain axis in bipolar depression. Mol. Psychiatry 2022, 27, 4123–4135. [Google Scholar] [CrossRef] [PubMed]
- Kootte, R.S.; Levin, E.; Salojärvi, J.; Smits, L.P.; Hartstra, A.V.; Udayappan, S.D.; Hermes, G.; Bouter, K.E.; Koopen, A.M.; Holst, J.J.; et al. Improvement of Insulin Sensitivity after Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition. Cell Metab. 2017, 26, 611–619.e6. [Google Scholar] [CrossRef] [Green Version]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef] [PubMed]
- Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C.; et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 2019, 4, 623–632. [Google Scholar] [CrossRef] [PubMed]
- Michels, N.; Van de Wiele, T.; De Henauw, S. Chronic Psychosocial Stress and Gut Health in Children: Associations with Calprotectin and Fecal Short-Chain Fatty Acids. Psychosom. Med. 2017, 79, 927–935. [Google Scholar] [CrossRef]
- Savignac, H.M.; Corona, G.; Mills, H.; Chen, L.; Spencer, J.P.; Tzortzis, G.; Burnet, P.W. Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-d-aspartate receptor subunits and d-serine. Neurochem. Int. 2013, 63, 756–764. [Google Scholar] [CrossRef] [Green Version]
- Qiu, X.; Wang, H.; Lan, Y.; Miao, J.; Pan, C.; Sun, W.; Li, G.; Wang, Y.; Zhao, X.; Zhu, Z.; et al. Blood biomarkers of post-stroke depression after minor stroke at three months in males and females. BMC Psychiatry 2022, 22, 162. [Google Scholar] [CrossRef]
- Vázquez, E.; Barranco, A.; Ramírez, M.; Gruart, A.; Delgado-García, J.M.; Martínez-Lara, E.; Blanco, S.; Martín, M.J.; Castanys, E.; Buck, R.; et al. Effects of a human milk oligosaccharide, 2′-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents. J. Nutr. Biochem. 2015, 26, 455–465. [Google Scholar] [CrossRef]
- Savignac, H.M.; Couch, Y.; Stratford, M.; Bannerman, D.M.; Tzortzis, G.; Anthony, D.C.; Burnet, P.W. Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice. Brain Behav. Immun. 2016, 52, 120–131. [Google Scholar] [CrossRef] [Green Version]
- O’Mahony, S.M.; Neufeld, K.-A.M.; Waworuntu, R.V.; Pusceddu, M.M.; Manurung, S.; Murphy, K.; Strain, C.; Laguna, M.C.; Peterson, V.L.; Stanton, C.; et al. The enduring effects of early-life stress on the microbiota–gut–brain axis are buffered by dietary supplementation with milk fat globule membrane and a prebiotic blend. Eur. J. Neurosci. 2020, 51, 1042–1058. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Spitzer, S.O.; Tkacz, A.; Savignac, H.M.; Cooper, M.; Giallourou, N.; Mann, E.O.; Bannerman, D.M.; Swann, J.R.; Anthony, D.C.; Poole, P.S.; et al. Postnatal prebiotic supplementation in rats affects adult anxious behaviour, hippocampus, electrophysiology, metabolomics, and gut microbiota. iScience 2021, 24, 103113. [Google Scholar] [CrossRef] [PubMed]
- Lehmann, S.; Hiller, J.; Van Bergenhenegouwen, J.; Knippels, L.M.J.; Garssen, J.; Traidl-Hoffmann, C. In Vitro Evidence for Immune-Modulatory Properties of Non-Digestible Oligosaccharides: Direct Effect on Human Monocyte Derived Dendritic Cells. PLoS ONE 2015, 10, e0132304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aoki, R.; Onuki, M.; Hattori, K.; Ito, M.; Yamada, T.; Kamikado, K.; Kim, Y.-G.; Nakamoto, N.; Kimura, I.; Clarke, J.M.; et al. Commensal microbe-derived acetate suppresses NAFLD/NASH development via hepatic FFAR2 signalling in mice. Microbiome 2021, 9, 188. [Google Scholar] [CrossRef]
- Belzung, C.; Griebel, G. Measuring normal and pathological anxiety-like behaviour in mice: A review. Behav. Brain Res. 2001, 125, 141–149. [Google Scholar] [CrossRef] [PubMed]
- Savignac, H.M.; Kiely, B.; Dinan, T.G.; Cryan, J.F. Bifidobacteriaexert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol. Motil. 2014, 26, 1615–1627. [Google Scholar] [CrossRef] [PubMed]
- Janik, R.; Thomason, L.A.; Stanisz, A.M.; Forsythe, P.; Bienenstock, J.; Stanisz, G.J. Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, N-acetyl aspartate and glutamate. Neuroimage 2016, 125, 988–995. [Google Scholar] [CrossRef]
- Dhaliwal, J.; Singh, D.P.; Singh, S.; Pinnaka, A.; Boparai, R.K.; Bishnoi, M.; Kondepudi, K.K.; Chopra, K. Lactobacillus plantarum MTCC 9510 supplementation protects from chronic unpredictable and sleep deprivation-induced behaviour, biochemical and selected gut microbial aberrations in mice. J. Appl. Microbiol. 2018, 125, 257–269. [Google Scholar] [CrossRef] [PubMed]
- Moya-Pérez, A.; Perez-Villalba, A.; Benítez-Páez, A.; Campillo, I.; Sanz, Y. Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice. Brain Behav. Immun. 2017, 65, 43–56. [Google Scholar] [CrossRef]
- Abildgaard, A.; Elfving, B.; Hokland, M.; Wegener, G.; Lund, S. Probiotic treatment reduces depressive-like behaviour in rats independently of diet. Psychoneuroendocrinology 2017, 79, 40–48. [Google Scholar] [CrossRef]
- Fachi, J.L.; Felipe, J.D.S.; Pral, L.P.; da Silva, B.K.; Corrêa, R.O.; de Andrade, M.C.P.; da Fonseca, D.M.; Basso, P.J.; Câmara, N.O.S.; Souza, L.D.S.E.; et al. Butyrate Protects Mice from Clostridium difficile-Induced Colitis through an HIF-1-Dependent Mechanism. Cell Rep. 2019, 27, 750–761.e7. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; Sun, J.; Wang, F.; Ding, G.; Chen, W.; Fang, R.; Yao, Y.; Pang, M.; Lu, Z.-Q.; Liu, J. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016, 1642, 70–78. [Google Scholar] [CrossRef]
- Berger, A. Science commentary: Th1 and Th2 responses: What are they? BMJ 2000, 321, 424. [Google Scholar] [CrossRef] [Green Version]
- Lugo-Huitrón, R.; Ugalde Muñiz, P.; Pineda, B.; Pedraza-Chaverrí, J.; Ríos, C.; Pérez-de La Cruz, V. Quinolinic acid: An endogenous neurotoxin with multiple targets. Oxidative Med. Cell. Longev. 2013, 2013, 104024. [Google Scholar] [CrossRef] [Green Version]
- Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.-C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, K.; Cowen, P.; Harmer, C.; Tzortzis, G.; Errington, S.; Burnet, P.W.J. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 2015, 232, 1793–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnstone, N.; Milesi, C.; Burn, O.; van den Bogert, B.; Nauta, A.; Hart, K.; Sowden, P.; Burnet, P.W.J.; Kadosh, K.C. Anxiolytic effects of a galacto-oligosaccharides prebiotic in healthy females (18–25 years) with corresponding changes in gut bacterial composition. Sci. Rep. 2021, 11, 8302. [Google Scholar] [CrossRef] [PubMed]
- Cho, H.S.; Park, J.M.; Lim, C.H.; Cho, Y.K.; Lee, I.S.; Kim, S.W.; Choi, M.-G.; Chung, I.-S.; Chung, Y.K. Anxiety, depression and quality of life in patients with irritable bowel syndrome. Gut Liver 2011, 5, 29–36. [Google Scholar] [CrossRef] [Green Version]
- Silk, D.B.A.; Davis, A.; Vulevic, J.; Tzortzis, G.; Gibson, G.R. Clinical trial: The effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2009, 29, 508–518. [Google Scholar] [CrossRef]
- Kazemi, A.; Noorbala, A.A.; Azam, K.; Eskandari, M.H.; Djafarian, K. Effect of probiotic and prebiotic vs. placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin. Nutr. 2019, 38, 522–528. [Google Scholar] [CrossRef] [PubMed]
- Liu, R.T.; Walsh, R.F.L.; Sheehan, A.E. Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci. Biobehav. Rev. 2019, 102, 13–23. [Google Scholar] [CrossRef]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
- Kelly, J.R.; Allen, A.P.; Temko, A.; Hutch, W.; Kennedy, P.J.; Farid, N.; Murphy, E.; Boylan, G.; Bienenstock, J.; Cryan, J.F.; et al. Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav. Immun. 2017, 61, 50–59. [Google Scholar] [CrossRef] [PubMed]
- Baião, R.; Capitão, L.P.; Higgins, C.; Browning, M.; Harmer, C.J.; Burnet, P.W.J. Multispecies probiotic administration reduces emotional salience and improves mood in subjects with moderate depression: A randomised, double-blind, placebo-controlled study. Psychol. Med. 2022, 1–11. [Google Scholar] [CrossRef]
- Romijn, A.R.; Rucklidge, J.J.; Kuijer, R.G.; Frampton, C. A double-blind, randomized, placebo-controlled trial of Lactobacillus helveticus and Bifidobacterium longum for the symptoms of depression. Aust. N. Z. J. Psychiatry 2017, 51, 810–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef] [PubMed]
- Browne, P.D.; Bolte, A.C.; der Vaart, I.B.-V.; Claassen, E.; de Weerth, C. Probiotics as a treatment for prenatal maternal anxiety and depression: A double-blind randomized pilot trial. Sci. Rep. 2021, 11, 3051. [Google Scholar] [CrossRef]
- Slykerman, R.F.; Hood, F.; Wickens, K.; Thompson, J.M.D.; Barthow, C.; Murphy, R.; Kang, J.; Rowden, J.; Stone, P.; Crane, J.; et al. Probiotic in Pregnancy Study Group. Effect of Lactobacillus rhamnosus HN001 in Pregnancy on Postpartum Symptoms of Depression and Anxiety: A Randomised Double-blind Placebo-controlled Trial. EBioMedicine 2017, 24, 159–165. [Google Scholar] [CrossRef] [Green Version]
- Dawe, J.P.; McCowan, L.M.E.; Wilson, J.; Okesene-Gafa, K.A.M.; Serlachius, A.S. Probiotics and Maternal Mental Health: A Randomised Controlled Trial among Pregnant Women with Obesity. Sci. Rep. 2020, 10, 1291. [Google Scholar] [CrossRef] [Green Version]
- Desai, V.; Kozyrskyj, A.L.; Lau, S.; Sanni, O.; Dennett, L.; Walter, J.; Ospina, M.B. Effectiveness of Probiotic, Prebiotic, and Synbiotic Supplementation to Improve Perinatal Mental Health in Mothers: A Systematic Review and Meta-Analysis. Front. Psychiatry 2021, 12, 622181. [Google Scholar] [CrossRef]
- Sohn, K.; Underwood, M.A. Prenatal and postnatal administration of prebiotics and probiotics. Semin. Fetal Neonatal Med. 2017, 22, 284–289. [Google Scholar] [CrossRef]
- Wickens, K.; Stanley, T.V.; Mitchell, E.A.; Barthow, C.; Fitzharris, P.; Purdie, G.; Siebers, R.; Black, P.N.; Crane, J. Early supplementation with Lactobacillus rhamnosus HN001 reduces eczema prevalence to 6 years: Does it also reduce atopic sensitization? Clin. Exp. Allergy 2013, 43, 1048–1057. [Google Scholar] [CrossRef]
- Wickens, K.; Barthow, C.; Mitchell, E.A.; Stanley, T.V.; Purdie, G.; Rowden, J.; Kang, J.; Hood, F.; van den Elsen, L.; Forbes-Blom, E.; et al. Maternal supplementation alone with Lactobacillus rhamnosus HN001 during pregnancy and breastfeeding does not reduce infant eczema. Pediatr. Allergy Immunol. 2018, 29, 296–302. [Google Scholar] [CrossRef] [PubMed]
- Barthow, C.; Wickens, K.; Stanley, T.; Mitchell, E.A.; Maude, R.; Abels, P.; Purdie, G.; Murphy, R.; Stone, P.; Kang, J.; et al. The Probiotics in Pregnancy Study (PiP Study): Rationale and design of a double-blind randomised controlled trial to improve maternal health during pregnancy and prevent infant eczema and allergy. BMC Pregnancy Childbirth 2016, 16, 133. [Google Scholar] [CrossRef] [Green Version]
- Baldassarre, M.E.; Palladino, V.; Amoruso, A.; Pindinelli, S.; Mastromarino, P.; Fanelli, M.; Di Mauro, A.; Laforgia, N. Rationale of Probiotic Supplementation during Pregnancy and Neonatal Period. Nutrients 2018, 10, 1693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hebert, J.C.; Radford-Smith, D.E.; Probert, F.; Ilott, N.; Chan, K.W.; Anthony, D.C.; Burnet, P.W.J. Mom’s diet matters: Maternal prebiotic intake in mice reduces anxiety and alters brain gene expression and the fecal microbiome in offspring. Brain Behav. Immun. 2021, 91, 230–244. [Google Scholar] [CrossRef] [PubMed]
- Williams, S.; Chen, L.; Savignac, H.M.; Tzortzis, G.; Anthony, D.; Burnet, P.W. Neonatal prebiotic (BGOS) supplementation increases the levels of synaptophysin, GluN2A-subunits and BDNF proteins in the adult rat hippocampus. Synapse 2016, 70, 121–124. [Google Scholar] [CrossRef]
- Han, M.-M.; Sun, J.-F.; Su, X.-H.; Peng, Y.-F.; Goyal, H.; Wu, C.-H.; Zhu, X.-Y.; Li, L. Probiotics improve glucose and lipid metabolism in pregnant women: A meta-analysis. Ann. Transl. Med. 2019, 7, 99. [Google Scholar] [CrossRef]
- Himaja, N.; Hemalatha, R.; Babu, K.N.; Shujauddin, M. Lactobacillus rhamnosus GG supplementation during critical windows of gestation influences immune phenotype in Swiss albino mice offspring. Benef. Microbes 2016, 7, 195–204. [Google Scholar] [CrossRef]
- Di Benedetto, M.G.; Bottanelli, C.; Cattaneo, A.; Pariante, C.M.; Borsini, A. Nutritional and immunological factors in breast milk: A role in the intergenerational transmission from maternal psychopathology to child development. Brain Behav. Immun. 2020, 85, 57–68. [Google Scholar] [CrossRef]
- Azagra-Boronat, I.; Tres, A.; Massot-Cladera, M.; Franch, À.; Castell, M.; Guardiola, F.; Pérez-Cano, F.J.; Rodríguez-Lagunas, M.J. Lactobacillus fermentum CECT5716 supplementation in rats during pregnancy and lactation affects mammary milk composition. J. Dairy Sci. 2020, 103, 2982–2992. [Google Scholar] [CrossRef]
- Azagra-Boronat, I.; Tres, A.; Massot-Cladera, M.; Franch, À.; Castell, M.; Guardiola, F.; Pérez-Cano, F.J.; Rodríguez-Lagunas, M.J. Lactobacillus fermentum CECT5716 Supplementation in Rats during Pregnancy and Lactation Impacts Maternal and Offspring Lipid Profile, Immune System and Microbiota. Cells 2020, 9, 575. [Google Scholar] [CrossRef]
- Radford-Smith, D.E.; Probert, F.; Burnet, P.W.J.; Anthony, D.C. Modifying the maternal microbiota alters the gut–brain metabolome and prevents emotional dysfunction in the adult offspring of obese dams. Proc. Natl. Acad. Sci. USA 2022, 119, e2108581119. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.-N.; Hou, C.-Y.; Chan, J.Y.; Lee, C.-T.; Tain, Y.-L. Hypertension Programmed by Perinatal High-Fat Diet: Effect of Maternal Gut Microbiota-Targeted Therapy. Nutrients 2019, 11, 2908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Radford-Smith, D.E.; Anthony, D.C. Mechanisms of Maternal Diet-Induced Obesity Affecting the Offspring Brain and Development of Affective Disorders. Metabolites 2023, 13, 455. [Google Scholar] [CrossRef] [PubMed]
- Buffington, S.A.; Di Prisco, G.V.; Auchtung, T.A.; Ajami, N.J.; Petrosino, J.F.; Costa-Mattioli, M. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016, 165, 1762–1775. [Google Scholar] [CrossRef] [Green Version]
- Ohland, C.L.; Kish, L.; Bell, H.; Thiesen, A.; Hotte, N.; Pankiv, E.; Madsen, K.L. Effects of Lactobacillus helveticus on murine behavior are dependent on diet and genotype and correlate with alterations in the gut microbiome. Psychoneuroendocrinology 2013, 38, 1738–1747. [Google Scholar] [CrossRef]
- Charbonneau, D.; Gibb, R.D.; Quigley, E.M. Fecal excretion of Bifidobacterium infantis 35624 and changes in fecal microbiota after eight weeks of oral supplementation with encapsulated probiotic. Gut Microbes 2013, 4, 201–211. [Google Scholar] [CrossRef] [Green Version]
- Bouhnik, Y.; Pochart, P.; Marteau, P.; Arlet, G.; Goderel, I.; Rambaud, J.C. Fecal recovery in humans of viable Bifidobacterium sp ingested in fermented milk. Gastroenterology 1992, 102, 875–878. [Google Scholar] [CrossRef]
- Maldonado-Gómez, M.X.; Martínez, I.; Bottacini, F.; O’Callaghan, A.; Ventura, M.; van Sinderen, D.; Hillmann, B.; Vangay, P.; Knights, D.; Hutkins, R.W.; et al. Stable Engraftment of Bifidobacterium longum AH1206 in the Human Gut Depends on Individualized Features of the Resident Microbiome. Cell Host Microbe 2016, 20, 515–526. [Google Scholar] [CrossRef] [Green Version]
- Dotterud, C.K.; Avershina, E.; Sekelja, M.; Simpson, M.; Rudi, K.; Storrø, O.; Johnsen, R.; Øien, T. Does Maternal Perinatal Probiotic Supplementation Alter the Intestinal Microbiota of Mother and Child? J. Pediatr. Gastroenterol. Nutr. 2015, 61, 200–207. [Google Scholar] [CrossRef]
- Schultz, M.; Göttl, C.; Young, R.J.; Iwen, P.; Vanderhoof, J.A. Administration of oral probiotic bacteria to pregnant women causes temporary infantile colonization. J. Craniofacial Surg. 2004, 38, 293–297. [Google Scholar] [CrossRef]
- Li, S.S.; Zhu, A.; Benes, V.; Costea, P.I.; Hercog, R.; Hildebrand, F.; Huerta-Cepas, J.; Nieuwdorp, M.; Salojärvi, J.; Voigt, A.Y.; et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 2016, 352, 586–589. [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] [Green Version]
- Oki, K.; Akiyama, T.; Matsuda, K.; Gawad, A.; Makino, H.; Ishikawa, E.; Oishi, K.; Kushiro, A.; Fujimoto, J. Long-term colonization exceeding six years from early infancy of Bifidobacterium longum subsp. longum in human gut. BMC Microbiol. 2018, 18, 209. [Google Scholar] [CrossRef] [PubMed]
- Yousuf, E.I.; Carvalho, M.; Dizzell, S.E.; Kim, S.; Gunn, E.; Twiss, J.; Giglia, L.; Stuart, C.; Hutton, E.K. Persistence of Suspected Probiotic Organisms in Preterm Infant Gut Microbiota Weeks after Probiotic Supplementation in the NICU. Front. Microbiol. 2020, 11, 574137. [Google Scholar] [CrossRef]
- Blüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef]
- Luppino, F.S.; de Wit, L.M.; Bouvy, P.F.; Stijnen, T.; Cuijpers, P.; Penninx, B.W.J.H.; Zitman, F.G. Overweight, Obesity, and Depression: A systematic review and meta-analysis of longitudinal studies. Arch. Gen. Psychiatry 2010, 67, 220–229. [Google Scholar] [CrossRef]
- Jung, S.J.; Woo, H.-T.; Cho, S.; Park, K.; Jeong, S.; Lee, Y.J.; Kang, D.; Shin, A. Association between body size, weight change and depression: Systematic review and meta-analysis. Br. J. Psychiatry 2017, 211, 14–21. [Google Scholar] [CrossRef] [Green Version]
- Pereira-Miranda, E.; Costa, P.R.; Queiroz, V.A.; Pereira-Santos, M.; Santana, M.L.P. Overweight and Obesity Associated with Higher Depression Prevalence in Adults: A Systematic Review and Meta-Analysis. J. Am. Coll. Nutr. 2017, 36, 223–233. [Google Scholar] [CrossRef]
- Radford-Smith, D.E.; Patel, P.J.; Irvine, K.M.; Russell, A.; Siskind, D.; Anthony, D.C.; Powell, E.E.; Probert, F. Depressive symptoms in non-alcoholic fatty liver disease are identified by perturbed lipid and lipoprotein metabolism. PLoS ONE 2022, 17, e0261555. [Google Scholar] [CrossRef]
- Fuller, N.R.; Burns, J.; Sainsbury, A.; Horsfield, S.; da Luz, F.; Zhang, S.; Denyer, G.; Markovic, T.P.; Caterson, I.D. Examining the association between depression and obesity during a weight management programme. Clin. Obes. 2017, 7, 354–359. [Google Scholar] [CrossRef]
- Gibson-Smith, D.; Bot, M.; Milaneschi, Y.; Twisk, J.W.; Visser, M.; Brouwer, I.; Penninx, B.W.J.H. Major depressive disorder, antidepressant use, and subsequent 2-Year weight change patterns in the Netherlands study of depression and anxiety. J. Clin. Psychiatry 2016, 77, e144–e151. [Google Scholar] [CrossRef] [PubMed]
- Wray, N.R.; Ripke, S.; Mattheisen, M.; Trzaskowski, M.; Byrne, E.M.; Abdellaoui, A.; Adams, M.J.; Agerbo, E.; Air, T.M.; Andlauer, T.M.F.; et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 2018, 50, 668–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Locke, A.E.; Kahali, B.; Berndt, S.I.; Justice, A.E.; Pers, T.H.; Day, F.R.; Powell, C.; Vedantam, S.; Buchkovich, M.L.; Yang, J.; et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 2015, 518, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thorleifsson, G.; Walters, G.B.; Gudbjartsson, D.F.; Steinthorsdottir, V.; Sulem, P.; Helgadottir, A.; Styrkarsdottir, U.; Gretarsdottir, S.; Thorlacius, S.; Jonsdottir, I.; et al. Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat. Genet. 2009, 41, 18–24. [Google Scholar] [CrossRef] [PubMed]
- Willer, C.J.; Speliotes, E.K.; Loos, R.J.F.; Li, S.; Lindgren, C.M.; Heid, I.M.; Berndt, S.I.; Elliott, A.L.; Jackson, A.U.; Lamina, C.; et al. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat. Genet. 2009, 41, 25–34. [Google Scholar] [CrossRef] [Green Version]
- Speliotes, E.K.; Willer, C.J.; Berndt, S.I.; Monda, K.L.; Thorleifsson, G.; Jackson, A.U.; Allen, H.L.; Lindgren, C.M.; Luan, J.; Mägi, R.; et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 2010, 42, 937–948. [Google Scholar] [CrossRef] [Green Version]
- The Early Growth Genetics (EGG) Consortium. A genome-wide association meta-analysis identifies new childhood obesity loci. Nat. Genet. 2012, 44, 526–531. [Google Scholar] [CrossRef]
- Berndt, S.I.; Gustafsson, S.; Mägi, R.; Ganna, A.; Wheeler, E.; Feitosa, M.F.; Justice, A.E.; Monda, K.L.; Croteau-Chonka, D.C.; Day, F.R.; et al. Genome-wide meta-analysis identifies 11 new loci for anthropometric traits and provides insights into genetic architecture. Nat. Genet. 2013, 45, 501–512. [Google Scholar] [CrossRef] [Green Version]
- Levey, D.F.; Stein, M.B.; Wendt, F.R.; Pathak, G.A.; Zhou, H.; Aslan, M.; Quaden, R.; Harrington, K.M.; Nuñez, Y.Z.; Overstreet, C.; et al. Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat. Neurosci. 2021, 24, 954–963. [Google Scholar] [CrossRef]
- Lee, A.W.S.; Hengstler, H.; Schwald, K.; Diaz, M.B.; Loreth, D.; Kirsch, M.; Kretz, O.; Haas, C.A.; de Angelis, M.H.; Herzig, S.; et al. Functional inactivation of the genome-wide association study obesity gene neuronal growth regulator 1 in mice causes a body mass phenotype. PLoS ONE 2012, 7, e41537. [Google Scholar] [CrossRef]
- Singh, K.; Jayaram, M.; Kaare, M.; Leidmaa, E.; Jagomäe, T.; Heinla, I.; Hickey, M.A.; Kaasik, A.; Schäfer, M.K.; Innos, J.; et al. Neural cell adhesion molecule Negr1 deficiency in mouse results in structural brain endophenotypes and behavioral deviations related to psychiatric disorders. Sci. Rep. 2019, 9, 5457. [Google Scholar] [CrossRef] [Green Version]
- Noh, K.; Lee, H.; Choi, T.-Y.; Joo, Y.; Kim, S.-J.; Kim, H.; Kim, J.Y.; Jahng, J.W.; Lee, S.; Choi, S.-Y.; et al. Negr1 controls adult hippocampal neurogenesis and affective behaviors. Mol. Psychiatry 2019, 24, 1189–1205. [Google Scholar] [CrossRef]
- Boender, A.J.; van Gestel, M.A.; Garner, K.M.; Luijendijk, M.C.M.; Adan, R.A.H. The obesity-associated gene Negr1 regulates aspects of energy balance in rat hypothalamic areas. Physiol. Rep. 2014, 2, e12083. [Google Scholar] [CrossRef] [Green Version]
- Milaneschi, Y.; Simmons, W.K.; Van Rossum, E.F.C.; Penninx, B.W. Depression and obesity: Evidence of shared biological mechanisms. Mol. Psychiatry 2019, 24, 18–33. [Google Scholar] [CrossRef]
- Bruce-Keller, A.J.; Salbaum, J.M.; Luo, M.; Blanchard, E.; Taylor, C.M.; Welsh, D.A.; Berthoud, H.-R. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol. Psychiatry 2015, 77, 607–615. [Google Scholar] [CrossRef] [Green Version]
- Vicentini, F.A.; Mathews, A.J.; Pittman, Q.J.; Swain, M.G.; Sharkey, K.A.; Hirota, S.A. Behavioural adaptations after antibiotic treatment in male mice are reversed by activation of the aryl hydrocarbon receptor. Brain Behav. Immun. 2021, 98, 317–329. [Google Scholar] [CrossRef]
- Bot, M.; Milaneschi, Y.; Al-Shehri, T.; Amin, N.; Garmaeva, S.; Onderwater, G.L.; Pool, R.; Thesing, C.S.; Vijfhuizen, L.S.; Vogelzangs, N.; et al. Metabolomics Profile in Depression: A Pooled Analysis of 230 Metabolic Markers in 5283 Cases with Depression and 10,145 Controls. Biol. Psychiatry 2020, 87, 409–418. [Google Scholar] [CrossRef]
- Zemdegs, J.; Martin, H.; Pintana, H.; Bullich, S.; Manta, S.; Marqués, M.A.; Moro, C.; Layé, S.; Ducrocq, F.; Chattipakorn, N.; et al. Metformin Promotes Anxiolytic and Antidepressant-Like Responses in Insulin-Resistant Mice by Decreasing Circulating Branched-Chain Amino Acids. J. Neurosci. 2019, 39, 5935–5948. [Google Scholar] [CrossRef] [Green Version]
- Chan, S.Y.; Probert, F.; Radford-Smith, D.E.; Hebert, J.C.; Claridge, T.D.W.; Anthony, D.C.; Burnet, P.W.J. Post-inflammatory behavioural despair in male mice is associated with reduced cortical glutamate-glutamine ratios, and circulating lipid and energy metabolites. Sci. Rep. 2020, 10, 16857. [Google Scholar] [CrossRef]
- Fernstrom, J.D. Large neutral amino acids: Dietary effects on brain neurochemistry and function. Amino Acids 2013, 45, 419–430. [Google Scholar] [CrossRef]
- Siddik, A.B.; Shin, A.C. Recent Progress on Branched-Chain Amino Acids in Obesity, Diabetes, and Beyond. Endocrinol. Metab. 2019, 34, 234–246. [Google Scholar] [CrossRef]
- Pietiläinen, K.H.; Naukkarinen, J.; Rissanen, A.; Saharinen, J.; Ellonen, P.; Keränen, H.; Suomalainen-Wartiovaara, A.; Götz, A.; Suortti, T.; Yki-Jarvinen, H.; et al. Global transcript profiles of fat in monozygotic twins discordant for BMI: Pathways behind acquired obesity. PLoS Med. 2008, 5, e51. [Google Scholar] [CrossRef]
- Mishra, P.K.; Kumar, A.; Behar, K.L.; Patel, A.B. Subanesthetic ketamine reverses neuronal and astroglial metabolic activity deficits in a social defeat model of depression. J. Neurochem. 2018, 146, 722–734. [Google Scholar] [CrossRef] [Green Version]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Radford-Smith, D.E.; Anthony, D.C. Prebiotic and Probiotic Modulation of the Microbiota–Gut–Brain Axis in Depression. Nutrients 2023, 15, 1880. https://doi.org/10.3390/nu15081880
Radford-Smith DE, Anthony DC. Prebiotic and Probiotic Modulation of the Microbiota–Gut–Brain Axis in Depression. Nutrients. 2023; 15(8):1880. https://doi.org/10.3390/nu15081880
Chicago/Turabian StyleRadford-Smith, Daniel E., and Daniel C. Anthony. 2023. "Prebiotic and Probiotic Modulation of the Microbiota–Gut–Brain Axis in Depression" Nutrients 15, no. 8: 1880. https://doi.org/10.3390/nu15081880
APA StyleRadford-Smith, D. E., & Anthony, D. C. (2023). Prebiotic and Probiotic Modulation of the Microbiota–Gut–Brain Axis in Depression. Nutrients, 15(8), 1880. https://doi.org/10.3390/nu15081880