Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2149 2024-01-04 20:19:30 |
2 format change + 1 word(s) 2150 2024-01-05 03:11:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Cheddadi, R.; Yeramilli, V.; Martin, C. Varied Microbiome Profiles in Neonates. Encyclopedia. Available online: https://encyclopedia.pub/entry/53448 (accessed on 19 May 2024).
Cheddadi R, Yeramilli V, Martin C. Varied Microbiome Profiles in Neonates. Encyclopedia. Available at: https://encyclopedia.pub/entry/53448. Accessed May 19, 2024.
Cheddadi, Riadh, Venkata Yeramilli, Colin Martin. "Varied Microbiome Profiles in Neonates" Encyclopedia, https://encyclopedia.pub/entry/53448 (accessed May 19, 2024).
Cheddadi, R., Yeramilli, V., & Martin, C. (2024, January 04). Varied Microbiome Profiles in Neonates. In Encyclopedia. https://encyclopedia.pub/entry/53448
Cheddadi, Riadh, et al. "Varied Microbiome Profiles in Neonates." Encyclopedia. Web. 04 January, 2024.
Varied Microbiome Profiles in Neonates
Edit

The field of human microbiome and gut microbial diversity research has witnessed a profound transformation, driven by advances in omics technologies. These advancements have unveiled essential connections between microbiome alterations and severe conditions, prompting the development of new frameworks through epidemiological studies. Traditionally, it was believed that each individual harbored unique microbial communities acquired early in life, evolving over the course of their lifetime, with little acknowledgment of any prenatal microbial development, but research challenges this belief. The neonatal microbiome’s onset, influenced by factors like delivery mode and maternal health, remains a subject of intense debate, hinting at potential intrauterine microbial processes. 

neonatal microbiome metabolites maternal microbiome microbiome transfer maternal influence pregnancy

1. Introduction

The field of human microbiome and gut microbial diversity research has undergone significant transformation in response to the rapid advancement of omics technologies. While various aspects of biological research have experienced considerable progress, the trajectory of microbiome investigations is distinctive. This transformation involves a transition from culture-based approaches for analyzing oral microbial composition to molecular techniques for identifying microbial profiles across diverse ecological niches within the human body [1][2]. These advancements have revealed crucial associations between microbiome alterations and severe conditions spanning from neuropsychiatric disorders to cancer. Consequently, the development of new frameworks and model systems has been facilitated by microbiome epidemiological studies [3][4]. As an illustration, a notable example is the demonstrated influence of the gut microbiome on modulating the response of melanoma to anti-programmed cell death 1 protein (PD-1) immunotherapy [5].
The prevailing notion was that each human harbored predominantly unique microbial communities, which were traditionally believed to be acquired in early life; this phenomenon has been the cornerstone of numerous surprises in both basic scientific research and translational applications, giving rise to multiple discoveries and an abundance of answers [6][7]. One highly debated facet of the microbiome is its temporal dynamics. Some advocate for the long-term stability of the human gut microbiota [8], while others illustrate a fluctuating array of microbiome profiles across various life stages [9]. A more plausible framework involves embracing both viewpoints, considering the microbiome as a persistent profile with relatively rapid transitions during specific critical periods. Microbial diversity varies significantly across diverse ecological niches within the adult human body.

2. The Neonatal Microbiome

The human gut microbiome exhibits remarkable richness and harbors dynamic populations of microorganisms, primarily characterized by the dominance of bacterial communities. This ecosystem comprises approximately 3.8 × 1013 cells, collectively bearing a genome that surpasses the human genome by approximately 150-fold [10]. The multitude of trillions of cells residing within the gut represents the most abundant microbial population, and their pivotal role in host development and overall health is mediated via direct interactions with the host or through the influence of various metabolites. These interactions occur within the context of a highly homeostatic ecosystem, often referred to as the host–symbiont or holobiont. This conceptual framework acknowledges the integral role of coevolution in shaping the composition of the gut microbiome and its impact on human development. Consequently, any disruption in the long- or short-term selective pressures acting on the microbiome is bound to have significant consequences for neonatal development [11][12][13].
The objective of this research is to establish a consensus regarding the onset of neonatal microbiome development (Figure 1). Historically, the prevailing view has held that the intrauterine environment is devoid of microorganisms, with gut colonization commencing only at the moment of birth [14]. Within this paradigm, researchers who have investigated the post-birth neonatal microbiome have portrayed it as an initial set of microbial sets that undergo maturation and progression into a more intricate microbiome, characterized by an enrichment of Bacteroides and Firmicutes, which are representative of an adult-like microbiome [15]. Within this perspective, the colonization of the neonatal gut constitutes a de novo construction of a microbial community and is influenced by a multitude of factors. These factors include considerations such as age, dietary regimen, method of delivery, concurrent health conditions, antibiotic usage, and the birthing environment of the infant (NICU) [16][17]. The relationship between neonatal age and the mode of birth presents an intriguing connection, given that a significant proportion of premature infants are delivered via a C-section [18]. It is important to note that the microbiota of preterm infants are observed to contribute to the maintenance of an already fragile innate immune system. Consequently, any aberrant colonization of the gut microbiota may lead to unfavorable outcomes [19].
Figure 1. An infant’s microbiome undergoes significant changes, both in terms of quantity and quality, due to various influences. These factors can be categorized into two main groups: those that occur before or after birth. Notably, the mother has a substantial impact on the prenatal development of the neonatal microbiome, and this influence persists even after birth through different mechanisms.

3. Microbial Transfer and Postnatal Influences

The concept of a placental microbiome remains “again” a subject of considerable debate [20]. Aagaard et al. reported the presence of a microbiome in the placenta, resembling that of the oral cavity, under sterile conditions [21]. Subsequent investigators have corroborated Aagaard’s findings and argued that if there is a microbiome in blood and other human niches previously deemed sterile, it logically follows that there should be a distinct microbiome in the placenta as well [21][22]. Another intriguing discovery from the same research project involved the investigation of a potential connection between specific placental microbiome types and neonatal outcomes. They identified distinct placental taxa that correlated with preterm birth, shedding light on a potential link between the placental microbiome and adverse pregnancy outcomes [21]. Conversely, other investigators have questioned this entity, and argued that the human placenta does not have a microbiome but it does represent a potential site for microbial acquisition. De Goffau et al. employed distinct methods for DNA extraction and detection, and they based their conclusions on several factors. One key factor was the notably low bacterial sequence biomass in DNA extracted from the placenta, with a significant portion attributed to potential contamination during labor, delivery, or laboratory processes [23][24].
The transmission of specific bacterial taxa is heavily dependent on the mode of birth. Infants either inherit a microbial package resembling that found in the mother’s birth canal, or this maternal–fetal microbial overlap is lost when neonates are delivered via a cesarean section [25]. In the case of a cesarean section, other factors appear to have a more significant influence on the postnatal microbiome as compared to the prenatal microbiome. These factors primarily include environmental elements such as delivery and surgical equipment, healthcare workers, and contact with other neonates [16]. Lactobacillus appears to be a prominent feature of vaginal delivery, being highly abundant, particularly in the maternal vagina. Infants born via cesarean section exhibited a consistently low detection rate of Lactobacilli for up to 6 months after birth. Surprisingly, this disparity in bacterial taxa disappears by the time the infants reach three years of age [26].
Similarly, medication, particularly antibiotics, has been implicated in causing deleterious effects on the human gut microbiome, particularly when administered during the early neonatal phase [27][28][29]. The microbial alterations induced by antibiotics are deemed to have adverse implications for the well-being and prospective development of newborns. Descriptively, antibiotic-triggered modifications to the microbiome appear to affect distinct facets, specifically, diversity, temporal stability, and quantitative distribution [30].
Environmental factors have also been documented to exert an influence on the neonatal microbiome. Studies have revealed disparities, particularly in terms of both the diversity and predominant bacterial types, contingent upon the birthplace. At the compositional level, infants born in hospital settings tend to exhibit lower levels of Bacteroides, Bifidobacterium, Streptococcus, and Lactobacillus, while displaying higher proportions of the Clostridium and Enterobacteriaceae families compared to infants born at home [31]. Notably, these disparities are reflective of a proinflammatory phenotype, marked by an overexpression of various inflammatory markers in infants born in hospital environments [32]. It is essential to emphasize, nevertheless, that some studies have demonstrated that the place of birth plays a role in the divergence of an initially similar microbiome. In other words, the initial bacterial colonization does not differ significantly between the two groups [33].

4. Prenatal Influences on Neonatal Microbiome

The concept of Developmental Origins of Health and Disease (DOHaD) encompasses a field of investigations proposing that detrimental exposures occurring in the early stages of life, while tissues and organs are undergoing development, may elevate the susceptibility to diseases later in life [34][35]. The majority of embryonic development and organogenesis happens during the intrauterine period, where the fertilized oocyte develops into a coordinated assembly of interconnected organs. In alignment with this paradigm, compelling epidemiological evidence and experimental data in animal models have substantiated a robust association between the intrauterine environment, often represented by the maternal factors, and the subsequent risk of infants developing diseases in later stages of life [36][37].
The concept of allostatic load finds its manifestation in the context of pregnancy, which can be regarded as a unique state of “allostasis” wherein the maternal–fetal dyad faces the intricate challenge of combining the dual objectives of maternal and fetal well-being across the course of development [38]. Maternal adversity experienced during pregnancy can be predominantly associated with environmental factors. The spectrum of maternal stressors is expansive and profoundly impactful, including factors such as partner violence, healthcare accessibility, housing conditions, experiences of humiliation, racial discrimination, and a heightened sense of danger. Many of these stressors can be comprehensively categorized under the broader umbrella of social determinants of health [39]. The mechanisms through which maternal adversity can influence microbiome development are interconnected with dysregulation of the hypothalamo-pituitary axis, pronounced cytokine secretion, direct placental effects, and metabolic alterations [40]. In a rodent model assessing stress during pregnancy, it has been illustrated that maternal stress exerts an influence on the postnatal colonic microbiome, manifesting from postnatal day 2 through postnatal day 28. Notably, a disrupted microbiome structure was detected in male offspring as they displayed characteristics akin to the microbiome patterns typically associated with females, thus suggesting a linkage between stress, hormonal factors, and the gut microbiome [41]. From a descriptive perspective, the stress-induced alterations in the microbiome encompassed a concurrent proliferation of facultative anaerobic microorganisms at the detriment of obligate anaerobes [42][43]. This observation is noteworthy, as it implies that specific taxa of facultative anaerobes, such as Mucispirillum and Desulfovibrionaceae, collectively possess the capacity for mucin degradation and the production of hydrogen sulfide [34]. These changes are regarded as a plausible foundation for the initiation of intestinal inflammation [44][45].
Maternal dietary patterns have the potential to impact the developing fetus through multiple mechanisms [46], and it is widely hypothesized that one of these mechanisms involves influencing the composition of the neonatal microbiome. A study investigating the microbial DNA in the amniotic fluid and placenta from pregnant mothers administered probiotic compounds and revealed a significant alteration in innate immunity gene patterns [47]. In this direction, specific probiotic bacterial-derived metabolites show promise as potential perinatal therapeutic interventions [48]. Numerous medications have been identified as having adverse effects on fetal development [49]. Nonetheless, only a limited number of drugs have been recognized for their ability to mitigate certain effects through the modulation of the maternal and fetal microbiome, with antibiotics being one notable example. In animal models, when administered to pregnant dams, it was noted that there were significant changes in the neonatal gut microbiome, a reduction in intestinal host defense, and an elevated risk of neonatal sepsis. These observations were attributed to a potential decrease in the transmission of bacteria during or shortly after delivery. It is also plausible, however, that maternal antibiotics may limit the colonization of the fetal gut by bacteria even before birth, leading to an atypical immune priming [50].

5. Interpreting Varied Microbiome Profiles

Up to this point, the researchers have discussed the factors contributing to a variable microbiome in neonates. The continuity involves pinpointing the relationships between specific microbiome taxa and particular health outcomes. It is imperative to acknowledge that an infant’s microbiota are inherently distinguished by lower bacterial abundance and diversity. As the infant matures, the microbiota progressively become more complex. Analogously, the rudimentary, less diverse microbiota in early infancy can be likened to a solid foundation for a building. Should this foundation be flawed, any structure constructed upon it is predisposed to instability and eventual deterioration [51][52][53]. Considering this perspective, particular microbiome profiles can be directly associated with specific diseases. In-depth research has demonstrated that disruptions in early-life microbiota are conducive to the development of obesity induced by a high-fat diet; further investigation showed that these alterations are primarily instigated by the depletion of Lactobacillus species within the gut microbiota [35]. In the same experimental model, it elucidated the decrease in Lactobacillus-derived metabolites, particularly phenyllactic acid, known to activate peroxisome proliferator-activated receptor γ (PPAR-γ), a key regulator of lipid metabolism [54]. Another illustration of how the microbiome engages with the immune system involves molecular signaling and the participation of innate immunity, facilitated by various microbial species and microbiome-related molecules. The colonization of the gut by Gram-negative bacteria that secrete lipopolysaccharides (LPSs) has been identified as a pivotal factor in this mechanism. LPS serves as the trigger for initiating inflammatory responses, particularly those mediated through Toll-like receptor 4 (TLR-4) and nuclear factor kappa B (NF-κB) [55][56][57].

References

  1. Costello, E.K.; Lauber, C.L.; Hamady, M.; Fierer, N.; Gordon, J.I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 1694–1697.
  2. The Integrative HMP (iHMP) Research Network Consortium. The Integrative Human Microbiome Project. Nature 2019, 569, 641–648.
  3. Wang, L.; Conlon, M.A.; Christophersen, C.T.; Sorich, M.J.; Angley, M.T. Gastrointestinal microbiota and metabolite biomarkers in children with autism spectrum disorders. Biomark Med. 2014, 8, 331–344.
  4. Ge, Y.; Wang, X.; Guo, Y.; Yan, J.; Abuduwaili, A.; Aximujiang, K.; Yan, J.; Wu, M. Gut microbiota influence tumor development and Alter interactions with the human immune system. J. Exp. Clin. Cancer Res. 2021, 40, 42.
  5. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103.
  6. Byrd, A.L.; Segre, J.A. Elucidating microbial codes to distinguish individuals. Proc. Natl. Acad. Sci. USA 2015, 112, 6778–6779.
  7. Franzosa, E.A.; Huang, K.; Meadow, J.F.; Gevers, D.; Lemon, K.P.; Bohannan, B.J.M.; Huttenhower, C. Identifying personal microbiomes using metagenomic codes. Proc. Natl. Acad. Sci. USA 2015, 112, E2930–E2938.
  8. Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The long-term stability of the human gut microbiota. Science 2013, 341, 1237439.
  9. Gajer, P.; Brotman, R.M.; Bai, G.; Sakamoto, J.; Schütte, U.M.E.; Zhong, X.; Koenig, S.S.K.; Fu, L.; Ma, Z.; Zhou, X.; et al. Temporal Dynamics of the Human Vaginal Microbiota. Sci. Transl. Med. 2012, 4, 132ra52.
  10. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533.
  11. Bordenstein, S.R.; Theis, K.R. Host Biology in Light of the Microbiome: Ten Principles of Holobionts and Hologenomes. PLoS Biol. 2015, 13, e1002226.
  12. Zilber-Rosenberg, I.; Rosenberg, E. Microbial-driven genetic variation in holobionts. FEMS Microbiol. Rev. 2021, 45, fuab022.
  13. Blanton, L.V.; Charbonneau, M.R.; Salih, T.; Barratt, M.J.; Venkatesh, S.; Ilkaveya, O.; Subramanian, S.; Manary, M.J.; Trehan, I.; Jorgensen, J.M.; et al. Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science 2016, 351, aad3311.
  14. Singh, A.; Mittal, M. Neonatal microbiome—A brief review. J. Matern. Fetal Neonatal. Med. 2020, 33, 3841–3848.
  15. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, O.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 852.
  16. Martin, R.; Makino, H.; Yavuz, A.C.; 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.
  17. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227.
  18. Thanh, B.Y.L.; Lumbiganon, P.; Pattanittum, P.; Laopaiboon, M.; Vogel, J.P.; Oladapo, O.T.; Pileggi-Castro, C.; Mori, R.; Jayaratne, K.; Qureshi, Z.; et al. Mode of delivery and pregnancy outcomes in preterm birth: A secondary analysis of the WHO Global and Multi-country Surveys. Sci. Rep. 2019, 9, 15556.
  19. Chong, C.Y.L.; Bloomfield, F.H.; O’Sullivan, J.M. Factors Affecting Gastrointestinal Microbiome Development in Neonates. Nutrients 2018, 10, 274.
  20. Dudley, D. The placental microbiome: Yea, nay or maybe? BJOG Int. J. Obstet. Gynaecol. 2020, 127, 170.
  21. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014, 6, 237ra65.
  22. D’Argenio, V. The Prenatal Microbiome: A New Player for Human Health. High Throughput 2018, 7, 38.
  23. de Goffau, M.C.; Lager, S.; Sovio, U.; Gaccioli, F.; Cook, E.; Peacock, S.J.; Parkhill, J.; Charnock-Jones, D.S.; Smith, G.C.S. Human placenta has no microbiome but can contain potential pathogens. Nature 2019, 572, 329–334.
  24. Sterpu, I.; Fransson, E.; Hugerth, L.W.; Du, J.; Pereira, M.; Cheng, L.; Radu, S.A.; Calderón-Pérez, L.; Zha, Y.; Angelidou, P.; et al. No evidence for a placental microbiome in human pregnancies at term. Am. J. Obstet. Gynecol. 2021, 224, 296.e1–296.e23.
  25. Jakobsson, H.E.; Abrahamsson, T.R.; Jenmalm, M.C.; Harris, K.H.; Quince, C.; Jernberg, C.; Björkstén, B.; Engstrand, L.; Andersson, A.F. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 2014, 63, 559–566.
  26. Nagpal, R.; Tsuji, H.; Takahashi, T.; Kawashima, K.; Nagata, S.; Nomoto, K.; Yamashiro, Y. Sensitive Quantitative Analysis of the Meconium Bacterial Microbiota in Healthy Term Infants Born Vaginally or by Cesarean Section. Front. Microbiol. 2016, 7, 1997.
  27. Gasparrini, A.J.; Wang, B.; Sun, X.; Kennedy, E.A.; Hernandez-Leyva, A.; Ndao, I.M.; Tarr, P.I.; Warner, B.B.; Dantas, G. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat. Microbiol. 2019, 4, 2285–2297.
  28. Russell, J.T.; Ruoss, J.L.; de la Cruz, D.; Li, N.; Bazacliu, C.; Patton, L.; McKinley, K.L.; Garrett, T.J.; Polin, R.A.; Triplett, E.W.; et al. Antibiotics and the developing intestinal microbiome, metabolome and inflammatory environment in a randomized trial of preterm infants. Sci. Rep. 2021, 11, 1943.
  29. Dardas, M.; Gill, S.R.; Grier, A.; Pryhuber, G.S.; Gill, A.L.; Lee, Y.-H.; Guillet, R. The impact of postnatal antibiotics on the preterm intestinal microbiome. Pediatr. Res. 2014, 76, 150–158.
  30. Cetinbas, M.; Thai, J.; Filatava, E.; Gregory, K.E.; Sadreyev, R.I. Long-term dysbiosis and fluctuations of gut microbiome in antibiotic treated preterm infants. iScience 2023, 26, 107995.
  31. Selma-Royo, M.; Arroyo, M.C.; García-Mantrana, I.; Parra-Llorca, A.; Escuriet, R.; Martínez-Costa, C.; Collado, M.C. Perinatal environment shapes microbiota colonization and infant growth: Impact on host response and intestinal function. Microbiome 2020, 8, 167.
  32. Combellick, J.L.; Shin, H.; Shin, D.; Cai, Y.; Hagan, H.; Lacher, C.; Lin, D.L.; McCauley, K.; Lynch, S.V.; Dominguez-Bello, M.G. Differences in the fecal microbiota of neonates born at home or in the hospital. Sci. Rep. 2018, 8, 15660.
  33. Stojanov, M.; Das, S.; Odent, M.; Engel, P.; Baud, D. Home or hospital birth: The neonatal microbiota perspective. Lancet Microbe 2022, 3, e247.
  34. Rooks, M.G.; Veiga, P.; Wardwell-Scott, L.-H.; Tickle, T.; Segata, N.; Michaud, M.; Gallini, C.A.; Beal, C.; van Hylckama-Vlieg, J.E.T.; Ballal, S.A.; et al. Gut microbiome composition and function in experimental colitis during active disease and treatment-induced remission. ISME J. 2014, 8, 1403–1417.
  35. Tohi, M.; Bay, J.L.; Tu’akoi, S.; Vickers, M.H. The Developmental Origins of Health and Disease: Adolescence as a Critical Lifecourse Period to Break the Transgenerational Cycle of NCDs-A Narrative Review. Int. J. Environ. Res Public Health 2022, 19, 6024.
  36. Khambadkone, S.G.; Cordner, Z.A.; Tamashiro, K.L.K. Maternal stressors and the developmental origins of neuropsychiatric risk. Front. Neuroendocrinol. 2020, 57, 100834.
  37. Fernandez-Twinn, D.S.; Hjort, L.; Novakovic, B.; Ozanne, S.E.; Saffery, R. Intrauterine programming of obesity and type 2 diabetes. Diabetologia 2019, 62, 1789–1801.
  38. Power, M.L.; Schulkin, J. Maternal obesity, metabolic disease, and allostatic load. Physiol. Behav. 2012, 106, 22–28.
  39. Cheddadi, R.; Khandekar, N.N.; Yeramilli, V.; Martin, C. The impact of maternal stress on the development of necrotizing enterocolitis: A comprehensive review. Semin. Pediatr. Surg. 2023, 32, 151324.
  40. Kramer, A.C.; Jansson, T.; Bale, T.L.; Powell, T.L. Maternal-fetal cross-talk via the placenta: Influence on offspring development and metabolism. Development 2023, 150(20), dev202088.
  41. Jašarević, E.; Howard, C.D.; Misic, A.M.; Beiting, D.P.; Bale, T.L. Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci. Rep. 2017, 7, 44182.
  42. Lyte, M.; Vulchanova, L.; Brown, D.R. Stress at the intestinal surface: Catecholamines and mucosa-bacteria interactions. Cell Tissue Res. 2011, 343, 23–32.
  43. Winter, S.E.; Bäumler, A.J. Why related bacterial species bloom simultaneously in the gut: Principles underlying the ‘Like will to like’ concept. Cell Microbiol. 2014, 16, 179–184.
  44. Robertson, B.R.; O’Rourke, J.L.; Neilan, B.A.; Vandamme, P.; On, S.L.W.; Fox, J.G.; Lee, A. Mucispirillum schaedleri gen. nov., sp. nov., a spiral-shaped bacterium colonizing the mucus layer of the gastrointestinal tract of laboratory rodents. Int. J. Syst. Evol. Microbiol. 2005, 55 Pt 3, 1199–1204.
  45. Berry, D.; Schwab, C.; Milinovich, G.; Reichert, J.; MahfoudhK, B.; Decker, T.; Engel, M.; Hai, B.; Hainzl, E.; Heider, S. Phylotype-level 16S rRNA analysis reveals new bacterial indicators of health state in acute murine colitis. ISME J. 2012, 6, 2091–2106.
  46. Blumfield, M.L.; Hure, A.J.; MacDonald-Wicks, L.K.; Smith, R.; Simpson, S.J.; Giles, W.B.; Raubenheimer, D.; Collins, C.E. Dietary balance during pregnancy is associated with fetal adiposity and fat distribution. Am. J. Clin. Nutr. 2012, 96, 1032–1041.
  47. Rautava, S.; Collado, M.C.; Salminen, S.; Isolauri, E. Probiotics modulate host-microbe interaction in the placenta and fetal gut: A randomized, double-blind, placebo-controlled trial. Neonatology 2012, 102, 178–184.
  48. Yan, F.; Cao, H.; Cover, T.L.; Whitehead, R.; Washington, M.K.; Polk, D.B. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007, 132, 562–575.
  49. Tsamantioti, E.S.; Hashmi, M.F. Teratogenic Medications. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023.
  50. Harshaw, C.; Kojima, S.; Wellman, C.L.; Demas, G.E.; Morrow, A.L.; Taft, D.H.; Kenkel, W.M.; Leffel, J.K.; Alberts, J.R. Maternal antibiotics disrupt microbiome, behavior, and temperature regulation in unexposed infant mice. Dev. Psychobiol. 2022, 64, e22289.
  51. Derrien, M.; Alvarez, A.-S.; de Vos, W.M. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019, 27, 997–1010.
  52. Roswall, J.; Olsson, L.M.; Kovatcheva-Datchary, P.; Nilsson, S.; Tremaroli, V.; Simon, M.-C.; Kiilerich, P.; Akrami, R.; Krämer, M.; Uhlén, M.; et al. Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe 2021, 29, 765–776.e3.
  53. Yeramilli, V.; Cheddadi, R.; Benjamin, H.; Martin, C. The Impact of Stress, Microbial Dysbiosis, and Inflammation on Necrotizing Enterocolitis. Microorganisms 2023, 11, 2206.
  54. Shelton, C.D.; Sing, E.; Mo, J.; Shealy, N.G.; Yoo, W.; Thomas, J.; Fitz, G.N.; Castro, P.R.; Hickman, T.T.; Torres, T.P.; et al. An early-life microbiota metabolite protects against obesity by regulating intestinal lipid metabolism. Cell Host Microbe 2023, 31, 1604–1619.e10.
  55. Pammi, M.; Hollister, E.; Neu, J. Gut Injury and the Microbiome in Neonates. Clin. Perinatol. 2020, 47, 369–382.
  56. Ng, P.C. An update on biomarkers of necrotizing enterocolitis. Semin. Fetal Neonatal Med. 2018, 23, 380–386.
  57. Wang, Y.; Jiang, K.; Xia, Q.; Kang, X.; Wang, S.; Yu, J.-H.; Ni, W.-F.; Qi, X.-Q.; Zhang, Y.-N.; Han, J.-B.; et al. Exploration of pathogenic microorganism within the small intestine of necrotizing enterocolitis. World J. Pediatr. 2023, 1–8.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 90
Revisions: 2 times (View History)
Update Date: 05 Jan 2024
1000/1000