Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life—A Narrative Review
Abstract
:1. Introduction
2. Materials and Methods
Literature Search
3. Microbiota and Immunity—Consequences of Gut Colonization
3.1. Effects on Innate Immunity Development
3.2. Effects on Adaptive Immunity Development
4. Microbiota and Cognition
- Hypothalamic–Pituitary–Adrenal (HPA) axis: A study by Gareau in 2014 discovered that germ-free mice exhibited an increase in HPA hyperactivity. Problems with the HPA axis in human gut dysbiosis can result in behaviors indicative of anxiety and depression [41].
- Neurotransmitters: The gut microbiome can modulate certain neurotransmitters, as seen with dopamine and serotonin production. Decreased levels of these neurotransmitters can directly affect a patient’s overall mood [45].
5. Immunity and Cognition
6. Discussion
6.1. Possible Gut-Immune-Cognition Interactions
6.2. Future Implications
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Wernroth, M.-L.; Peura, S.; Hedman, A.M.; Hetty, S.; Vicenzi, S.; Kennedy, B.; Fall, K.; Svennblad, B.; Andolf, E.; Pershagen, G.; et al. Development of gut microbiota during the first 2 years of life. Sci. Rep. 2022, 12, 9080. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; 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]
- Casella, G.; Tontini, G.E.; Bassotti, G.; Pastorelli, L.; Villanacci, V.; Spina, L.; Baldini, V.; Vecchi, M. Neurological disorders and inflammatory bowel diseases. World J. Gastroenterol. 2014, 20, 8764–8782. [Google Scholar] [CrossRef] [PubMed]
- Camilleri, M. Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut 2019, 68, 1516–1526. [Google Scholar] [CrossRef] [PubMed]
- Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schafer, D.P.; Stevens, B. Microglia Function in Central Nervous System Development and Plasticity. Cold Spring Harb. Perspect. Biol. 2015, 7, a020545. [Google Scholar] [CrossRef]
- McGuckin, M.A.; Lindén, S.K.; Sutton, P.; Florin, T.H. Mucin dynamics and enteric pathogens. Nat. Rev. Genet. 2011, 9, 265–278. [Google Scholar] [CrossRef]
- Bevins, C.L.; Salzman, N.H. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nat. Rev. Genet. 2011, 9, 356–368. [Google Scholar] [CrossRef]
- Cerutti, A.; Rescigno, M. The Biology of Intestinal Immunoglobulin A Responses. Immunity 2008, 28, 740–750. [Google Scholar] [CrossRef] [Green Version]
- Wagner, C.; Wheeler, K.; Ribbeck, K. Mucins and Their Role in Shaping the Functions of Mucus Barriers. Annu. Rev. Cell Dev. Biol. 2018, 34, 189–215. [Google Scholar] [CrossRef] [Green Version]
- Goto, Y.; Uematsu, S.; Kiyono, H. Epithelial glycosylation in gut homeostasis and inflammation. Nat. Immunol. 2016, 17, 1244–1251. [Google Scholar] [CrossRef] [PubMed]
- Johansson, M.E.; Jakobsson, H.E.; Holmén-Larsson, J.; Schütte, A.; Ermund, A.; Rodríguez-Piñeiro, A.M.; Arike, L.; Wising, C.; Svensson, F.; Bäckhed, F.; et al. Normalization of Host Intestinal Mucus Layers Requires Long-Term Microbial Colonization. Cell Host Microbe 2015, 18, 582–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padra, M.; Adamczyk, B.; Flahou, B.; Erhardsson, M.; Chahal, G.; Smet, A.; Jin, C.; Thorell, A.; Ducatelle, R.; Haesebrouck, F.; et al. Helicobacter suis infection alters glycosylation and decreases the pathogen growth inhibiting effect and binding avidity of gastric mucins. Mucosal Immunol. 2019, 12, 784–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venkatakrishnan, V.; Quintana-Hayashi, M.P.; Mahu, M.; Haesebrouck, F.; Pasmans, F.; Lindén, S.K. Brachyspira hyodysenteriae Infection Regulates Mucin Glycosylation Synthesis Inducing an Increased Expression of Core-2 O-Glycans in Porcine Colon. J. Proteome Res. 2017, 16, 1728–1742. [Google Scholar] [CrossRef]
- Vaishnava, S.; Behrendt, C.L.; Ismail, A.S.; Eckmann, L.; Hooper, L.V. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc. Natl. Acad. Sci. USA 2008, 105, 20858–20863. [Google Scholar] [CrossRef] [Green Version]
- Kespohl, M.; Vachharajani, N.; Luu, M.; Harb, H.; Pautz, S.; Wolff, S.; Sillner, N.; Walker, A.; Schmitt-Kopplin, P.; Boettger, T.; et al. The Microbial Metabolite Butyrate Induces Expression of Th1-Associated Factors in CD4+ T Cells. Front. Immunol. 2017, 8, 1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, W.; Sun, M.; Chen, F.; Cao, A.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol. 2017, 10, 946–956. [Google Scholar] [CrossRef] [Green Version]
- Macia, L.; Tan, J.; Vieira, A.T.; Leach, K.; Stanley, D.; Luong, S.; Maruya, M.; McKenzie, C.l.; Hijikata, A.; Wong, C.; et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 2015, 6, 6734. [Google Scholar] [CrossRef] [Green Version]
- Elce, A.; Amato, F.; Zarrilli, F.; Calignano, A.; Troncone, R.; Castaldo, G.; Canani, R.B. Butyrate modulating effects on pro-inflammatory pathways in human intestinal epithelial cells. Benef. Microbes 2017, 8, 841–847. [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]
- Li, Y.; Ye, Z.; Zhu, J.; Fang, S.; Meng, L.; Zhou, C. Effects of Gut Microbiota on Host Adaptive Immunity Under Immune Homeostasis and Tumor Pathology State. Front. Immunol. 2022, 13, 844335. [Google Scholar] [CrossRef] [PubMed]
- Davis, C.P.; Savage, D.C. Habitat, Succession, Attachment, and Morphology of Segmented, Filamentous Microbes Indigenous to the Murine Gastrointestinal Tract. Infect Immun. 1974, 10, 948–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.M.; Donaldson, G.P.; Mikulski, Z.; Boyajian, S.; Ley, K.; Mazmanian, S.K. Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 2013, 501, 426–429. [Google Scholar] [CrossRef] [Green Version]
- Zaborin, A.; Penalver Bernabe, B.; Keskey, R.; Sangwan, N.; Hyoju, S.; Gottel, N.; Gilbert, J.A.; Zaborina, O.; Alverdy, J.C. Spatial Compartmentalization of the Microbiome between the Lumen and Crypts Is Lost in the Murine Cecum following the Process of Surgery, Including Overnight Fasting and Exposure to Antibiotics. mSystems 2020, 5, e00377-20. [Google Scholar] [CrossRef]
- Mabbott, N.A.; Donaldson, D.S.; Ohno, H.; Williams, I.R.; Mahajan, A. Microfold (M) cells: Important immunosurveillance posts in the intestinal epithelium. Mucosal Immunol. 2013, 6, 666–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rescigno, M.; Urbano, M.; Valzasina, B.; Francolini, M.; Rotta, G.; Bonasio, R.; Granucci, F.; Kraehenbuhl, J.-P.; Ricciardi-Castagnoli, P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat. Immunol. 2001, 2, 361–367. [Google Scholar] [CrossRef] [PubMed]
- Kraehenbuhl, J.-P.; Corbett, M. Keeping the Gut Microflora at Bay. Science 2004, 303, 1624–1625. [Google Scholar] [CrossRef] [PubMed]
- Bunker, J.J.; Flynn, T.M.; Koval, J.C.; Shaw, D.G.; Meisel, M.; McDonald, B.D.; Ishizuka, I.E.; Dent, A.L.; Wilson, P.C.; Jabri, B.; et al. Innate and Adaptive Humoral Responses Coat Distinct Commensal Bacteria with Immunoglobulin A. Immunity 2015, 43, 541–553. [Google Scholar] [CrossRef] [Green Version]
- Uematsu, S.; Fujimoto, K.; Jang, M.H.; Yang, B.-G.; Jung, Y.-J.; Nishiyama, M.; Sato, S.; Tsujimura, T.; Yamamoto, M.; Yokota, Y.; et al. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat. Immunol. 2008, 9, 769–776. [Google Scholar] [CrossRef]
- Macpherson, A.J.; Gatto, D.; Sainsbury, E.; Harriman, G.R.; Hengartner, H.; Zinkernagel, R.M. A Primitive T Cell-Independent Mechanism of Intestinal Mucosal IgA Responses to Commensal Bacteria. Science 2000, 288, 2222–2226. [Google Scholar] [CrossRef]
- Johansen, F.-E.; Kaetzel, C.S. Regulation of the polymeric immunoglobulin receptor and IgA transport: New advances in environmental factors that stimulate pIgR expression and its role in mucosal immunity. Mucosal Immunol. 2011, 4, 598–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hapfelmeier, S.; Lawson, M.A.; Slack, E.; Kirundi, J.K.; Stoel, M.; Heikenwalder, M.; Cahenzli, J.; Velykoredko, Y.; Balmer, M.L.; Endt, K.; et al. Reversible Microbial Colonization of Germ-Free Mice Reveals the Dynamics of IgA Immune Responses. Science 2010, 328, 1705–1709. [Google Scholar] [CrossRef] [Green Version]
- Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular Analysis of Commensal Host-Microbial Relationships in the Intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [Green Version]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [Green Version]
- Sharon, G.; Cruz, N.J.; Kang, D.-W.; Gandal, M.J.; Wang, B.; Kim, Y.-M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; Lane, C.J.; et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019, 177, 1600–1618.e17. [Google Scholar] [CrossRef] [Green Version]
- Carlson, A.L.; Xia, K.; Azcarate-Peril, M.A.; Goldman, B.D.; Ahn, M.; Styner, M.A.; Thompson, A.L.; Geng, X.; Gilmore, J.H.; Knickmeyer, R.C. Infant Gut Microbiome Associated with Cognitive Development. Biol. Psychiatry 2018, 83, 148–159. [Google Scholar] [CrossRef] [PubMed]
- Tomova, A.; Husarova, V.; Lakatosova, S.; Bakos, J.; Vlkova, B.; Babinska, K.; Ostatnikova, D. Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav. 2015, 138, 179–187. [Google Scholar] [CrossRef] [PubMed]
- Sittipo, P.; Choi, J.; Lee, S.; Lee, Y.K. The function of gut microbiota in immune-related neurological disorders: A review. J. Neuroinflamm. 2022, 19, 154. [Google Scholar] [CrossRef]
- Proctor, C.; Thiennimitr, P.; Chattipakorn, N.; Chattipakorn, S.C. Diet, gut microbiota and cognition. Metab. Brain Dis. 2017, 32, 1–17. [Google Scholar] [CrossRef]
- Gareau, M.G.; Wine, E.; Rodrigues, D.M.; Cho, J.H.; Whary, M.T.; Philpott, D.J.; MacQueen, G.; Sherman, P.M. Bacterial infection causes stress-induced memory dysfunction in mice. Gut 2011, 60, 307–317. [Google Scholar] [CrossRef]
- Gareau, M.G. Microbiota-Gut-Brain Axis and Cognitive Function; Springer: New York, NY, USA, 2014; Volume 817, pp. 357–371. [Google Scholar] [CrossRef]
- Gao, W.; Baumgartel, K.L.; Alexander, S.A. The Gut Microbiome as a Component of the Gut–Brain Axis in Cognitive Health. Biol. Res. Nurs. 2020, 22, 485–494. [Google Scholar] [CrossRef] [PubMed]
- Carlson, A.L.; Xia, K.; Azcarate-Peril, M.A.; Rosin, S.P.; Fine, J.P.; Mu, W.; Zopp, J.B.; Kimmel, M.C.; Styner, M.A.; Thompson, A.L.; et al. Infant gut microbiome composition is associated with non-social fear behavior in a pilot study. Nat. Commun. 2021, 12, 3294. [Google Scholar] [CrossRef] [PubMed]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
- Matsumoto, M.; Kibe, R.; Ooga, T.; Aiba, Y.; Sawaki, E.; Koga, Y.; Benno, Y. Cerebral Low-Molecular Metabolites Influenced by Intestinal Microbiota: A Pilot Study. Front. Syst. Neurosci. 2013, 7, 9. [Google Scholar] [CrossRef] [Green Version]
- Lu, J.; Synowiec, S.; Lu, L.; Yu, Y.; Bretherick, T.; Takada, S.; Yarnykh, V.; Caplan, J.; Caplan, M.; Claud, E.C.; et al. Microbiota influence the development of the brain and behaviors in C57BL/6J mice. PLoS ONE 2018, 13, e0201829. [Google Scholar] [CrossRef]
- Calcia, M.A.; Bonsall, D.R.; Bloomfield, P.S.; Selvaraj, S.; Barichello, T.; Howes, O.D. Stress and neuroinflammation: A systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology 2016, 233, 1637–1650. [Google Scholar] [CrossRef] [Green Version]
- Hoeijmakers, L.; Lucassen, P.J.; Korosi, A. The interplay of early-life stress, nutrition, and immune activation programs adult hippocampal structure and function. Front. Mol. Neurosci. 2015, 7, 103. [Google Scholar] [CrossRef]
- Miller, G.E.; Chen, E.; Fok, A.K.; Walker, H.; Lim, A.; Nicholls, E.F.; Cole, S.; Kobor, M.S. Low early-life social class leaves a biological residue manifested by decreased glucocorticoid and increased proinflammatory signaling. Proc. Natl. Acad. Sci. USA 2009, 106, 14716–14721. [Google Scholar] [CrossRef] [Green Version]
- Deykin, E.Y.; Macmahon, B. VIRAL EXPOSURE AND AUTISM. Am. J. Epidemiol. 1979, 109, 628–638. [Google Scholar] [CrossRef]
- Chess, S.; Fernandez, P.; Korn, S. Behavioral consequences of congenital rubella. J. Pediatr. 1978, 93, 699–703. [Google Scholar] [CrossRef]
- Goines, P.; van de Water, J. The immune system’s role in the biology of autism. Curr. Opin. Neurol. 2010, 23, 111–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ricci, S.; Businaro, R.; Ippoliti, F.; Vasco, V.R.L.; Massoni, F.; Onofri, E.; Troili, G.M.; Pontecorvi, V.; Morelli, M.; Ricciardi, M.R.; et al. Altered Cytokine and BDNF Levels in Autism Spectrum Disorder. Neurotox. Res. 2013, 24, 491–501. [Google Scholar] [CrossRef] [PubMed]
- Vargas, D.L.; Nascimbene, C.; Krishnan, C.; Zimmerman, A.W.; Pardo, C.A. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann. Neurol. 2005, 57, 67–81. [Google Scholar] [CrossRef]
- Wei, H.; Chadman, K.K.; McCloskey, D.P.; Sheikh, A.M.; Malik, M.; Brown, W.T.; Li, X. Brain IL-6 elevation causes neuronal circuitry imbalances and mediates autism-like behaviors. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2012, 1822, 831–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cusick, S.E.; Georgieff, M.K. The Role of Nutrition in Brain Development: The Golden Opportunity of the “First 1000 Days”. J. Pediatr. 2016, 175, 16–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- John, C.C.; Black, M.M.; Nelson, C.A. Neurodevelopment: The Impact of Nutrition and Inflammation During Early to Middle Childhood in Low-Resource Settings. Pediatrics 2017, 139 (Suppl. S1), S59–S71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nuriel-Ohayon, M.; Neuman, H.; Koren, O. Microbial Changes during Pregnancy, Birth, and Infancy. Front. Microbiol. 2016, 7, 1031. [Google Scholar] [CrossRef] [Green Version]
- Aziz, Q.; Doré, J.; Emmanuel, A.; Guarner, F.; Quigley, E.M.M. Gut microbiota and gastrointestinal health: Current concepts and future directions. Neurogastroenterol. Motil. 2013, 25, 4–15. [Google Scholar] [CrossRef]
- Yong, E. Gut microbial ‘enterotypes’ become less clear-cut. Nature 2012, 10. [Google Scholar] [CrossRef]
- Penders, J.; Thijs, C.; Vink, C.; Stelma, F.F.; Snijders, B.; Kummeling, I.; Van den Brandt, P.A.; Stobberingh, E.E. Factors Influencing the Composition of the Intestinal Microbiota in Early Infancy. Pediatrics 2006, 118, 511–521. [Google Scholar] [CrossRef] [Green Version]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bentley, J.P.; Simpson, J.M.; Bowen, J.R.; Morris, J.M.; Roberts, C.L.; Nassar, N. Gestational age, mode of birth and breastmilk feeding all influence acute early childhood gastroenteritis: A record-linkage cohort study. BMC Pediatr. 2016, 16, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, R.; Makino, H.; Cetinyurek Yavuz, A.; Ben-Amor, K.; Roelofs, M.; Ishikawa, E.; Kubota, H.; Swinkels, S.; Sakai, T.; Oishi, K.; et al. Early-Life Events, Including Mode of Delivery and Type of Feeding, Siblings and Gender, Shape the Developing Gut Microbiota. PLoS ONE 2016, 11, e0158498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rutayisire, E.; Huang, K.; Liu, Y.; Tao, F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: A systematic review. BMC Gastroenterol. 2016, 16, 86. [Google Scholar] [CrossRef] [Green Version]
- Yasmin, F.; Tun, H.M.; Konya, T.B.; Guttman, D.S.; Chari, R.S.; Field, C.J.; Becker, A.B.; Mandhane, P.J.; Turvey, S.E.; Subbarao, P.; et al. Cesarean section, formula feeding, and infant antibiotic exposure: Separate and combined impacts on gut microbial changes in later infancy. Front. Pediatr. 2017, 10, 200. [Google Scholar] [CrossRef] [PubMed]
- Fouhy, F.; Watkins, C.; Hill, C.J.; O’Shea, C.A.; Nagle, B.; Dempsey, E.M.; O’Toole, P.W.; Ross, R.P.; Ryan, C.A.; Stanton, C. Perinatal factors affect the gut microbiota up to four years after birth. Nat. Commun. 2019, 10, 1517. [Google Scholar] [CrossRef] [Green Version]
- Trehan, I.; Goldbach, H.S.; LaGrone, L.N.; Meuli, G.J.; Wang, R.J.; Maleta, K.M.; Manary, M.J. Antibiotics as Part of the Management of Severe Acute Malnutrition. N. Engl. J. Med. 2013, 368, 425–435. [Google Scholar] [CrossRef] [Green Version]
- Kuperman, A.A.; Koren, O. Antibiotic use during pregnancy: How bad is it? BMC Med. 2016, 14, 91. [Google Scholar] [CrossRef] [Green Version]
- Gough, E.K.; Moodie, E.E.M.; Prendergast, A.; Johnson, S.M.A.; Humphrey, J.H.; Stoltzfus, R.J.; Walker, A.S.; Trehan, I.; Gibb, D.M.; Goto, R.; et al. The impact of antibiotics on growth in children in low and middle income countries: Systematic review and meta-analysis of randomised controlled trials. BMJ 2014, 348, g2267. [Google Scholar] [CrossRef] [Green Version]
- Guarner, F.; Malagelada, J.-R. Gut flora in health and disease. Lancet 2003, 361, 512–519. [Google Scholar] [CrossRef]
- Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019, 7, e7502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yassour, M.; Vatanen, T.; Siljander, H.; Hämäläinen, A.M.; Härkönen, T.; Ryhänen, S.J.; Franzosa, E.A.; Vlamakis, H.; Huttenhower, C.; Gevers, D.; et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016, 8, 343ra81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bratu, S.; Eramo, A.; Kopec, R.; Coughlin, E.; Ghitan, M.; Yost, R.; Chapnick, E.K.; Landman, D.; Quale, J. Community-associated Methicillin-resistant Staphylococcus aureus in Hospital Nursery and Maternity Units. Emerg. Infect. Dis. 2005, 11, 808–813. [Google Scholar] [CrossRef] [PubMed]
- Backhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Supporting data for the dynamics and stabilization of the Human gut microbiome during the first year of life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef] [Green Version]
- Azad, M.B.; Konya, T.; Maughan, H.; Guttman, D.S.; Field, C.J.; Chari, R.S.; Sears, M.R.; Becker, A.B.; Scott, J.A.; Kozyrskyj, A.L. Gut microbiota of healthy Canadian infants: Profiles by mode of delivery and infant diet at 4 months. Can. Med. Assoc. J. 2013, 185, 385–394. [Google Scholar] [CrossRef] [Green Version]
- Laursen, M.F. Gut Microbiota Development: Influence of Diet from Infancy to Toddlerhood. Ann. Nutr. Metab. 2021, 77, 21–34. [Google Scholar] [CrossRef]
- Duranti, S.; Lugli, G.A.; Mancabelli, L.; Armanini, F.; Turroni, F.; James, K.; Ferretti, P.; Gorfer, V.; Ferrario, C.; Milani, C.; et al. Maternal inheritance of bifidobacterial communities and bifidophages in infants through vertical transmission. Microbiome 2017, 5, 66. [Google Scholar] [CrossRef]
- Martín, R.; Jiménez, E.; Heilig, H.; Fernández, L.; Marín, M.L.; Zoetendal, E.G.; Rodríguez, J.M. Isolation of Bifidobacteria from Breast Milk and Assessment of the Bifidobacterial Population by PCR-Denaturing Gradient Gel Electrophoresis and Quantitative Real-Time, P.C.R. Appl. Environ. Microbiol. 2009, 75, 965–969. [Google Scholar] [CrossRef] [Green Version]
- Turroni, F.; Milani, C.; Duranti, S.; Mancabelli, L.; Mangifesta, M.; Viappiani, A.; Lugli, G.A.; Ferrario, C.; Gioiosa, L.; Ferrarini, A.; et al. Deciphering bifidobacterial-mediated metabolic interactions and their impact on gut microbiota by a multi-omics approach. ISME J. 2016, 10, 1656–1668. [Google Scholar] [CrossRef]
- Egan, M.; O’Connell Motherway, M.; Kilcoyne, M.; Kane, M.; Joshi, L.; Ventura, M.; Van Sinderen, D. Cross-feeding by Bifidobacterium breve UCC2003 during co-cultivation with Bifidobacterium bifidum PRL2010 in a mucin-based medium. BMC Microbiol. 2014, 14, 282. [Google Scholar] [CrossRef] [Green Version]
- Xiong, X.; Loo, S.L.; Zhang, L.; Tanaka, M.M. Modelling the effect of birth and feeding modes on the development of human gut microbiota. Proc. R. Soc. B Biol. Sci. 2021, 288, 20201810. [Google Scholar] [CrossRef] [PubMed]
- Mantis, N.J.; Forbes, S.J. Secretory IgA: Arresting Microbial Pathogens at Epithelial Borders. Immunol. Investig. 2010, 39, 383–406. [Google Scholar] [CrossRef] [PubMed]
- Lundell, A.-C.; Björnsson, V.; Ljung, A.; Ceder, M.; Johansen, S.; Lindhagen, G.; Törnhage, C.-J.; Adlerberth, I.; Wold, A.E.; Rudin, A. Infant B Cell Memory Differentiation and Early Gut Bacterial Colonization. J. Immunol. 2012, 188, 4315–4322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thompson, A.L.; Monteagudo-Mera, A.; Cadenas, M.B.; Lampl, M.L.; Azcarate-Peril, M.A. Milk- and solid-feeding practices and daycare attendance are associated with differences in bacterial diversity, predominant communities, and metabolic and immune function of the infant gut microbiome. Front. Cell. Infect. Microbiol. 2015, 5, 3. [Google Scholar] [CrossRef] [Green Version]
- Differding, M.K.; Benjamin-Neelon, S.E.; Hoyo, C.; Østbye, T.; Mueller, N.T. Timing of complementary feeding is associated with gut microbiota diversity and composition and short chain fatty acid concentrations over the first year of life. BMC Microbiol. 2020, 20, 56. [Google Scholar] [CrossRef] [Green Version]
- Leong, C.; Haszard, J.J.; Lawley, B.; Otal, A.; Taylor, R.W.; Szymlek-Gay, E.A.; Fleming, E.A.; Daniels, L.; Fangupo, L.J.; Tannock, G.W.; et al. Mediation Analysis as a Means of Identifying Dietary Components That Differentially Affect the Fecal Microbiota of Infants Weaned by Modified Baby-Led and Traditional Approaches. Appl. Environ. Microbiol. 2018, 84, e00914-18. [Google Scholar] [CrossRef] [Green Version]
- Grier, A.; Qiu, X.; Bandyopadhyay, S.; Holden-Wiltse, J.; Kessler, H.A.; Gill, A.L.; Hamilton, B.; Huyck, H.; Misra, S.; Mariani, T.J.; et al. Impact of prematurity and nutrition on the developing gut microbiome and preterm infant growth. Microbiome 2017, 5, 158. [Google Scholar] [CrossRef]
- Walker, R.W.; Clemente, J.C.; Peter, I.; Loos, R.J.F. The prenatal gut microbiome: Are we colonized with bacteria in utero? Pediatr. Obes. 2017, 12, 3–17. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.A.; Jen, R.; Butler, A.; Lavoie, P.M. The developing human preterm neonatal immune system: A case for more research in this area. Clin. Immunol. 2012, 145, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collado, M.C.; Cernada, M.; Neu, J.; Pérez-Martínez, G.; Gormaz, M.; Vento, M. Factors influencing gastrointestinal tract and microbiota immune interaction in preterm infants. Pediatr. Res. 2015, 77, 726–731. [Google Scholar] [CrossRef] [PubMed]
- Sherman, M.P. New Concepts of Microbial Translocation in the Neonatal Intestine: Mechanisms and Prevention. Clin. Perinatol. 2010, 37, 565–579. [Google Scholar] [CrossRef] [Green Version]
- Yoo, J.; Groer, M.; Dutra, S.; Sarkar, A.; McSkimming, D. Gut Microbiota and Immune System Interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef] [PubMed]
- Weng, M.; Walker, W.A. The role of gut microbiota in programming the immune phenotype. J. Dev. Orig. Heal. Dis. 2013, 4, 203–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Croxford, A.L.; Spath, S.; Becher, B. GM-CSF in Neuroinflammation: Licensing Myeloid Cells for Tissue Damage. Trends Immunol. 2015, 36, 651–662. [Google Scholar] [CrossRef]
- Jay, T.; Miller, C.M.; Cheng-Hathaway, P.; Graham, L.C.; BeMiller, S.; Broihier, M.L.; Xu, G.; Margevicius, D.; Karlo, J.C.; Sousa, G.; et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J. Exp. Med. 2015, 212, 287–295. [Google Scholar] [CrossRef]
- Gadani, S.P.; Walsh, J.T.; Lukens, J.R.; Kipnis, J. Dealing with Danger in the CNS: The Response of the Immune System to Injury. Neuron 2015, 87, 47–62. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Dissing-Olesen, L.; MacVicar, B.A.; Stevens, B. Microglia: Dynamic mediators of synapse development and plasticity. Trends Immunol. 2015, 36, 605–613. [Google Scholar] [CrossRef] [Green Version]
- Clarke, B.L.; Bost, K.L. Differential expression of functional adrenocorticotropic hormone receptors by subpopulations of lymphocytes. J Immunol. 1989, 143, 464–469. [Google Scholar] [CrossRef]
- Blalock, J.E. Proopiomelanocortin and the Immune-Neuroendocrine Connection. Ann. N. Y. Acad. Sci. 2006, 885, 161–172. [Google Scholar] [CrossRef] [PubMed]
- Stein, C.; Clark, J.D.; Oh, U.; Vasko, M.R.; Wilcox, G.L.; Overland, A.C.; Vanderah, T.W.; Spencer, R.H. Peripheral mechanisms of pain and analgesia. Brain Res. Rev. 2009, 60, 90–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zengeler, K.E.; Lukens, J.R. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat. Rev. Immunol. 2021, 21, 454–468. [Google Scholar] [CrossRef] [PubMed]
- Marı́n-Teva, J.L.; Dusart, I.; Colin, C.; Gervais, A.; van Rooijen, N.; Mallat, M. Microglia Promote the Death of Developing Purkinje Cells. Neuron 2004, 41, 535–547. [Google Scholar] [CrossRef] [PubMed]
- Lim, S.-H.; Park, E.; You, B.; Jung, Y.; Park, A.-R.; Park, S.G.; Lee, J.-R. Neuronal Synapse Formation Induced by Microglia and Interleukin 10. PLoS ONE 2013, 8, e81218. [Google Scholar] [CrossRef] [Green Version]
- Zhan, Y.; Paolicelli, R.C.; Sforazzini, F.; Weinhard, L.; Bolasco, G.; Pagani, F.; Vyssotski, A.L.; Bifone, A.; Gozzi, A.; Ragozzino, D.; et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat. Neurosci. 2014, 17, 400–406. [Google Scholar] [CrossRef]
- Gandal, M.J.; Haney, J.R.; Parikshak, N.N.; Leppa, V.; Ramaswami, G.; Hartl, C.; Schork, A.J.; Appadurai, V.; Buil, A.; Werge, T.M.; et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science 2018, 359, 693–697. [Google Scholar] [CrossRef] [Green Version]
- Dantzer, R. Neuroimmune Interactions: From the Brain to the Immune System and Vice Versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef]
- Borsini, A.; Zunszain, P.A.; Thuret, S.; Pariante, C.M. The role of inflammatory cytokines as key modulators of neurogenesis. Trends Neurosci. 2015, 38, 145–157. [Google Scholar] [CrossRef] [Green Version]
- Ma, L.; Li, X.W.; Zhang, S.J.; Yang, F.; Zhu, G.M.; Yuan, X.B.; Jiang, W. Interleukin-1 beta guides the migration of cortical neurons. J. Neuroinflamm. 2014, 11, 114. [Google Scholar] [CrossRef] [Green Version]
- Gadani, S.P.; Cronk, J.C.; Norris, G.T.; Kipnis, J. IL-4 in the Brain: A Cytokine to Remember. J. Immunol. 2012, 189, 4213–4219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vainchtein, I.D.; Chin, G.; Cho, F.S.; Kelley, K.W.; Miller, J.G.; Chien, E.C.; Liddelow, S.A.; Nguyen, P.T.; Nakao-Inoue, H.; Dorman, L.C.; et al. Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 2018, 359, 1269–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate Mapping Analysis Reveals That Adult Microglia Derive from Primitive Macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salvo-Romero, E.; Stokes, P.; Gareau, M.G. Microbiota-immune interactions: From gut to brain. LymphoSign J. 2020, 7, 1–23. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the, CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
- Bercik, P.; Denou, E.; Collins, J.; Jackson, W.; Lu, J.; Jury, J.; Deng, Y.; Blennerhassett, P.; Macri, J.; McCoy, K.D.; et al. The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology 2011, 141, 599–609.e3. [Google Scholar] [CrossRef] [Green Version]
- Milo, L.; Reardon, K.; Tappenden, K. Effects of short-chain fatty acid-supplemented total parenteral nutrition on intestinal pro-inflammatory cytokine abundance. Dig. Dis. Sci. 2002, 47, 2049–2055. [Google Scholar] [CrossRef]
- Korn, T.; Anderson, A.C.; Bettelli, E.; Oukka, M. The dynamics of effector T cells and Foxp3+ regulatory T cells in the promotion and regulation of autoimmune encephalomyelitis. J. Neuroimmunol. 2007, 191, 51–60. [Google Scholar] [CrossRef] [Green Version]
- Haghikia, A.; Jörg, S.; Duscha, A.; Berg, J.; Manzel, A.; Waschbisch, A.; Hammer, A.; Lee, D.H.; May, C.; Wilck, N.; et al. Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 2015, 43, 817–829. [Google Scholar] [CrossRef] [Green Version]
- Möhle, L.; Mattei, D.; Heimesaat, M.M.; Bereswill, S.; Fischer, A.; Alutis, M.; French, T.; Hambardzumyan, D.; Matzinger, P.; Dunay, I.R.; et al. Ly6Chi Monocytes Provide a Link between Antibiotic-Induced Changes in Gut Microbiota and Adult Hippocampal Neurogenesis. Cell. Rep. 2016, 15, 1945–1956. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Sanctuary, M.R.; Kain, J.N.; Chen, S.Y.; Kalanetra, K.; Lemay, D.G.; Rose, D.R.; Yang, H.T.; Tancredi, D.J.; German, J.B.; Slupsky, C.M.; et al. Pilot study of probiotic/colostrum supplementation on gut function in children with autism and gastrointestinal symptoms. PLoS ONE 2019, 14, e0210064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deleemans, J.M.; Chleilat, F.; Reimer, R.A.; Henning, J.-W.; Baydoun, M.; Piedalue, K.-A.; McLennan, A.; Carlson, L.E. The chemo-gut study: Investigating the long-term effects of chemotherapy on gut microbiota, metabolic, immune, psychological and cognitive parameters in young adult Cancer survivors; study protocol. BMC Cancer 2019, 19, 1243. [Google Scholar] [CrossRef] [Green Version]
- Jordan, K.R.; Loman, B.R.; Bailey, M.T.; Pyter, L.M. Gut microbiota-immune-brain interactions in chemotherapy-associated behavioral comorbidities. Cancer 2018, 124, 3990–3999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seitz, D.C.; Besier, T.; Debatin, K.-M.; Grabow, D.; Dieluweit, U.; Hinz, A.; Kaatsch, P.; Goldbeck, L. Posttraumatic stress, depression and anxiety among adult long-term survivors of cancer in adolescence. Eur. J. Cancer 2010, 46, 1596–1606. [Google Scholar] [CrossRef]
- Montassier, E.; Gastinne, T.; Vangay, P.; Al-Ghalith, G.A.; Bruley des Varannes, S.; Massart, S.; Moreau, P.; Potel, G.; De La Cochetière, M.F.; Batard, E.; et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment. Pharmacol. Ther. 2015, 42, 515–528. [Google Scholar] [CrossRef] [Green Version]
- Labus, J.S.; Hollister, E.B.; Jacobs, J.; Kirbach, K.; Oezguen, N.; Gupta, A.; Acosta, J.; Luna, R.A.; Aagaard, K.; Versalovic, J.; et al. Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome. Microbiome 2017, 5, 49. [Google Scholar] [CrossRef] [Green Version]
- Sim, K.; Shaw, A.G.; Randell, P.; Cox, M.J.; McClure, Z.E.; Li, M.-S.; Haddad, M.; Langford, P.R.; Cookson, W.O.C.M.; Moffatt, M.F.; et al. Dysbiosis Anticipating Necrotizing Enterocolitis in Very Premature Infants. Clin. Infect. Dis. 2015, 60, 389–397. [Google Scholar] [CrossRef] [Green Version]
- Johnson, C.C.; Ownby, D.R. The infant gut bacterial microbiota and risk of pediatric asthma and allergic diseases. Transl. Res. 2017, 179, 60–70. [Google Scholar] [CrossRef] [Green Version]
- Fujimura, K.E.; Sitarik, A.R.; Havstad, S.; Lin, D.L.; Levan, S.; Fadrosh, D.; Panzer, A.R.; LaMere, B.; Rackaityte, E.; Lukacs, N.W.; et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 2016, 22, 1187–1191. [Google Scholar] [CrossRef] [Green Version]
- Kalliomäki, M.; Collado, M.C.; Salminen, S.; Isolauri, E. Early differences in fecal microbiota composition in children may predict overweight. Am. J. Clin. Nutr. 2008, 87, 534–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buono, J.L.; Carson, R.T.; Flores, N.M. Health-related quality of life, work productivity, and indirect costs among patients with irritable bowel syndrome with diarrhea. Heal. Qual. Life Outcomes 2017, 15, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sandler, R.S.; Everhart, J.E.; Donowitz, M.; Adams, E.; Cronin, K.; Goodman, C.; Gemmen, E.; Shah, S.; Avdic, A.; Rubin, R. The burden of selected digestive diseases in the United States. Gastroenterology 2002, 122, 1500–1511. [Google Scholar] [CrossRef] [PubMed]
- Longstreth, G.F.; Wilson, A.; Knight, K.; Wong, J.; Chiou, C.F.; Barghout, V.; Frech, F.; Ofman, J.J. Irritable Bowel Syndrome, Health Care Use, and Costs: A U.S. Managed Care Perspective. Am. J. Gastroenterol. 2003, 98, 600–607. [Google Scholar] [CrossRef] [PubMed]
- Amaruddin, A.I.; Hamid, F.; Koopman, J.P.R.; Muhammad, M.; Brienen, E.A.; van Lieshout, L.; Geelen, A.R.; Wahyuni, S.; Kuijper, E.J.; Sartono, E.; et al. The Bacterial Gut Microbiota of Schoolchildren from High and Low Socioeconomic Status: A Study in an Urban Area of Makassar, Indonesia. Microorganisms 2020, 8, 961. [Google Scholar] [CrossRef]
- Surono, I.S.; Widiyanti, D.; Kusumo, P.D.; Venema, K. Gut microbiota profile of Indonesian stunted children and children with normal nutritional status. PLoS ONE 2021, 16, e0245399. [Google Scholar] [CrossRef] [PubMed]
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
Kartjito, M.S.; Yosia, M.; Wasito, E.; Soloan, G.; Agussalim, A.F.; Basrowi, R.W. Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life—A Narrative Review. Nutrients 2023, 15, 2642. https://doi.org/10.3390/nu15122642
Kartjito MS, Yosia M, Wasito E, Soloan G, Agussalim AF, Basrowi RW. Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life—A Narrative Review. Nutrients. 2023; 15(12):2642. https://doi.org/10.3390/nu15122642
Chicago/Turabian StyleKartjito, Melissa Stephanie, Mikhael Yosia, Erika Wasito, Garry Soloan, Achmad Furqan Agussalim, and Ray Wagiu Basrowi. 2023. "Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life—A Narrative Review" Nutrients 15, no. 12: 2642. https://doi.org/10.3390/nu15122642
APA StyleKartjito, M. S., Yosia, M., Wasito, E., Soloan, G., Agussalim, A. F., & Basrowi, R. W. (2023). Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life—A Narrative Review. Nutrients, 15(12), 2642. https://doi.org/10.3390/nu15122642