1
|
Mikhalev SA, Kurtser MA, Radzinsky VE, Orazov MR, Beeraka NM, Mikhaleva LM. Exploring the Role of Lower Genital Tract Microbiota and Cervical-Endometrial Immune Metabolome in Unknown Genesis of Recurrent Pregnancy Loss. Int J Mol Sci 2025; 26:1326. [PMID: 39941094 PMCID: PMC11818274 DOI: 10.3390/ijms26031326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Recurrent pregnancy loss (RPL) of unknown genesis is a complex condition with multifactorial origins, including genetic, hormonal, and immunological factors. However, the specific mechanisms underlying endocervical cell proliferation disorders in women with RPL remain inadequately understood, particularly concerning the role of microbiota and viral infections. The aim of this study was to investigate the mechanisms of endocervical cell proliferation disorders in women with RPL of unknown genesis by examining microbiota, human papillomavirus (HPV) typing, and the expression levels of key molecular biological markers, including p16/Ki-67, BCL-2, miR-145, and miR-34a. A prospective observational comparative study was executed on women with RPL and healthy pregnant controls with full ethical approval. Samples were collected for HPV typing and immunocytochemical analysis to evaluate the expression of p16, Ki-67, BCL-2, and the anti-oncogenic microRNAs (miR-145 and miR-34a). The expression of mRNA for the progesterone receptor (PGR-A) was also assessed, alongside local immune status markers, including proinflammatory T-lymphocytes (Th17/Th1) and regulatory CD4+ Tregs. Overexpression of p16, Ki-67, and BCL-2 was observed in 52.5% of women with RPL who had an ASC-US/LSIL cytogram, with the average double expression of p16/Ki-67 being three times higher than in the healthy pregnant group. A significant decrease in PGR-A mRNA expression in the endocervix of women with RPL was noted, accompanied by a dysregulated local immune status characterized by an increased prevalence of Th17/Th1 cells and a reduction in regulatory CD4+ Tregs. Additionally, the expression of miR-145 and miR-34a in the endocervix and endometrium of women with RPL significantly differed from the physiological pregnancy group, particularly in the context of high-risk HPV infection. The findings describe that disorders of endocervical cell proliferation in women with RPL of unknown genesis are associated with overexpression of specific molecular markers, impaired immune regulation, and altered microRNA profiles. These alterations may contribute to the pathophysiology of RPL, highlighting the need for further research into targeted interventions that could improve reproductive outcomes in affected individuals.
Collapse
Affiliation(s)
- Sergey A. Mikhalev
- Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov Russian National Research Medical University” of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (S.A.M.); (M.A.K.)
- City Clinical Hospital No. 31 Named After Academician G.M. Savelyeva of the Department of Health, 119415 Moscow, Russia
| | - Mark A. Kurtser
- Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov Russian National Research Medical University” of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (S.A.M.); (M.A.K.)
| | - Victor E. Radzinsky
- Department of Obstetrics and Gynecology, Federal State Autonomous Educational Institution of Higher Education «Peoples’ Friendship University of Russia», 117198 Moscow, Russia; (V.E.R.); (M.R.O.)
| | - Mekan R. Orazov
- Department of Obstetrics and Gynecology, Federal State Autonomous Educational Institution of Higher Education «Peoples’ Friendship University of Russia», 117198 Moscow, Russia; (V.E.R.); (M.R.O.)
| | - Narasimha M. Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu 515721, Andhra Pradesh, India
- Department of Studies in Molecular Biology, University of Mysore, Mysore 570006, Karnataka, India
| | - Lyudmila M. Mikhaleva
- Scientific Research Institute of Human Morphology Named After Academician A.P. Avtsyn of the Federal State Budgetary Scientific Institution “Russian Scientific Center of Surgery Named After Academician B.V. Petrovsky”, 125315 Moscow, Russia
| |
Collapse
|
2
|
Faas MM, Smink AM. Shaping immunity: the influence of the maternal gut bacteria on fetal immune development. Semin Immunopathol 2025; 47:13. [PMID: 39891756 PMCID: PMC11787218 DOI: 10.1007/s00281-025-01039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
The development of the fetal immune response is a highly complex process. In the present review, we describe the development of the fetal immune response and the role of the maternal gut bacteria in this process. In contrast to the previous belief that the fetal immune response is inert, it is now thought that the fetal immune response is uniquely tolerant to maternal and allo-antigens, but able to respond to infectious agents, such as bacteria. This is accomplished by the development of T cells toward regulatory T cells rather than toward effector T cells, but also by the presence of functional innate immune cells, such as monocytes and NK cells. Moreover, in fetuses there is different programming of CD8 + T cells and memory T cells toward innate immune cells rather than to adaptive immune cells. The maternal gut bacteria are important in shaping the fetal immune response by producing bacterial products and metabolites that pass the placenta into the fetus and influence development of the fetal immune response. Insight into how and when these products affect the fetal immune response may open new treatment options with pre- or probiotics to affect the maternal gut bacteria and therewith the fetal immune response.
Collapse
Affiliation(s)
- Marijke M Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands.
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands
| |
Collapse
|
3
|
Lapolla A, Dalfrà MG, Marelli G, Parrillo M, Sciacca L, Sculli MA, Succurro E, Torlone E, Vitacolonna E. Medical nutrition therapy in physiological pregnancy and in pregnancy complicated by obesity and/or diabetes: SID-AMD recommendations. Acta Diabetol 2025:10.1007/s00592-024-02442-7. [PMID: 39841216 DOI: 10.1007/s00592-024-02442-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025]
Abstract
Proper nutrition is essential during pregnancy to ensure an adequate supply of nutrients to the foetus and adequate maternal weight gain. In pregnancy complicated by diabetes (both gestational and pre-gestational), diet in terms of both the intake and quality of carbohydrates is an essential factor in glycaemic control. Maternal BMI at conception defines the correct weight increase during gestation in order to reduce maternal-foetal complications related to hypo- or hyper-nutrition. The recommendations presented here, which are based on national and international guidelines and the most recently published data on nutrition in physiological pregnancy and pregnancy complicated by hyperglycaemia and/or obesity, are designed to help healthcare professionals prescribe suitable eating patterns to safeguard the health of the mother and the foetus.
Collapse
Affiliation(s)
| | | | - Giuseppe Marelli
- Ordine Ospedaliero San Giovanni di Dio Fatebenefratelli, Erba, CO, Italy
| | - Mario Parrillo
- UOSD Endocrinologia e Malattie del Ricambio, AO Sant'Anna e San Sebastiano, Caserta, Italy
| | - Laura Sciacca
- Dipartimento Medicina Clinica e Sperimentale, Università degli Studi di Catania, Catania, Italy
| | - Maria Angela Sculli
- UOC Diabetologia e Endocrinologia, GOM Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Elena Succurro
- DPT Scienze Mediche Chirurgiche, Università Magna Grecia, Catanzaro, Italy
| | - Elisabetta Torlone
- AOS Maria della Misericordia SC Endocrinologia e Metabolismo, Università di Perugia, Perugia, Italy
| | - Ester Vitacolonna
- Dipartimento di Medicina e Scienza dell'Invecchiamento, Università di Chieti, Chieti, Italy
| |
Collapse
|
4
|
Masson BA, Kiridena P, Lu D, Kleeman EA, Reisinger SN, Qin W, Davies WJ, Muralitharan RR, Jama HA, Antonacci S, Marques FZ, Gubert C, Hannan AJ. Depletion of the paternal gut microbiome alters sperm small RNAs and impacts offspring physiology and behavior in mice. Brain Behav Immun 2025; 123:290-305. [PMID: 39293692 DOI: 10.1016/j.bbi.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiome on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that they may be relevant to paternal immune activation (PIA) modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiome. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles and offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.
Collapse
Affiliation(s)
- Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamudika Kiridena
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - William J Davies
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Simona Antonacci
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Sáez-Fuertes L, Kapravelou G, Grases-Pintó B, Massot-Cladera M, Bernabeu M, Knipping K, Garssen J, Bourdet-Sicard R, Castell M, Rodríguez-Lagunas MJ, Collado MC, Pérez-Cano FJ. Impact of maternal Bifidobacterium breve M-16V and scGOS/lcFOS supplementation during pregnancy and lactation on the maternal immune system and milk composition. Front Immunol 2024; 15:1418594. [PMID: 38975337 PMCID: PMC11224147 DOI: 10.3389/fimmu.2024.1418594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Maternal synbiotic supplementation during pregnancy and lactation can significantly influence the immune system. Prebiotics and probiotics have a positive impact on the immune system by preventing or ameliorating among others intestinal disorders. This study focused on the immunomodulatory effects of B. breve M-16V and short chain galacto-oligosaccharides (scGOS)/long chain fructo-oligosachairdes (lcFOS), including systemic and mucosal compartments and milk composition. Methods Lewis rats were orally administered with the synbiotic or vehicle during pregnancy (21 days) and lactation (21 days). At the weaning day, small intestine (SI), mammary gland (MG), adipose tissue, milk, mesenteric lymph nodes (MLN), salivary gland (SG), feces and cecal content were collected from the mothers. Results The immunoglobulinome profile showed increased IgG2c in plasma and milk, as well as elevated sIgA in feces at weaning. The supplementation improved lipid metabolism through enhanced brown adipose tissue activity and reinforced the intestinal barrier by increasing the expression of Muc3, Cldn4, and Ocln. The higher production of short chain fatty acids in the cecum and increased Bifidobacterium counts suggest a potential positive impact on the gastrointestinal tract. Discussion These findings indicate that maternal synbiotic supplementation during gestation and lactation improves their immunological status and improved milk composition.
Collapse
Affiliation(s)
- Laura Sáez-Fuertes
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Garyfallia Kapravelou
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
| | - Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Malen Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Manuel Bernabeu
- Institute of Agrochemisty and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Karen Knipping
- Division Immunology, Danone Nutricia Research, Utrecht, Netherlands
| | - Johan Garssen
- Division Immunology, Danone Nutricia Research, Utrecht, Netherlands
- Division Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Raphaëlle Bourdet-Sicard
- Life Science and Digital Health, Danone Global Research & Innovation Center, Gif-sur-Yvette, France
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
- Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Salud Carlos III, Madrid, Spain
| | - Maria José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| | - Maria Carmen Collado
- Institute of Agrochemisty and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Francisco José Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA-UB), Santa Coloma de Gramenet, Spain
| |
Collapse
|
6
|
Malloy E, Kates AE, Dixon J, Riley C, Safdar N, Hanson L. Vaginal and Rectal microbiome changes following administration of a multi-species antenatal probiotic: A randomized control trial. GUT MICROBES REPORTS 2024; 1:1-10. [PMID: 38708373 PMCID: PMC11065196 DOI: 10.1080/29933935.2024.2334311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/07/2024]
Abstract
The gut and vaginal microbiome undergo changes during pregnancy which may be protective or harmful to the birthing person. Probiotics have been found to cause protective changes to the gut and vaginal microbiomes, with the potential to improve perinatal outcomes. This randomized control trial compares the vaginal and rectal microbiomes before and after an antenatal probiotic or placebo intervention, with a diverse group of pregnant people and a special focus on racial disparities. The vaginal and rectal microbiomes reveal non-significant increased Lactobacillus in the probiotics group, with a greater increase in participants who identified as Black. Potential implications and future study are discussed.
Collapse
Affiliation(s)
- Emily Malloy
- Aurora UW Medical Group Midwifery & Wellness, Advocate Aurora Healthcare Milwaukee, USA
- College of Nursing, Marquette University, Milwaukee, USA
| | - Ashley E. Kates
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | - Jonah Dixon
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | - Colleen Riley
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | - Nasia Safdar
- Department of Medicine, Division of Infectious Disease, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | - Lisa Hanson
- College of Nursing, Marquette University, Milwaukee, USA
| |
Collapse
|
7
|
Tzitiridou-Chatzopoulou M, Kazakos E, Orovou E, Andronikidi PE, Kyrailidi F, Mouratidou MC, Iatrakis G, Kountouras J. The Role of Helicobacter pylori and Metabolic Syndrome-Related Mast Cell Activation Pathologies and Their Potential Impact on Pregnancy and Neonatal Outcomes. J Clin Med 2024; 13:2360. [PMID: 38673633 PMCID: PMC11050948 DOI: 10.3390/jcm13082360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Helicobacter pylori infection, a significant global burden beyond the gastrointestinal tract, has long been implicated in various systemic pathologies. Rising evidence suggests that the bacterium's intricate relationship with the immune system and its potential to induce chronic inflammation impact diverse pathophysiological processes in pregnant women that may in turn affect the incidence of several adverse pregnancy and neonate outcomes. Helicobacter pylori infection, which has been linked to metabolic syndrome and other disorders by provoking pericyte dysfunction, hyperhomocysteinemia, galectin-3, atrial fibrillation, gut dysbiosis, and mast cell activation pathologies, may also contribute to adverse pregnancy and neonatal outcomes. Together with increasing our biological understanding of the individual and collective involvement of Helicobacter pylori infection-related metabolic syndrome and concurrent activation of mast cells in maternal, fetus, and neonatal health outcomes, the present narrative review may foster related research endeavors to offer novel therapeutic approaches and informed clinical practice interventions to mitigate relevant risks of this critical topic among pregnant women and their offspring.
Collapse
Affiliation(s)
- Maria Tzitiridou-Chatzopoulou
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Evangelos Kazakos
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Eirini Orovou
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
| | - Paraskevi Eva Andronikidi
- Department of Nephrology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Maria C. Mouratidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Georgios Iatrakis
- Department of Midwifery, University of West Attica, 12243 Athens, Greece;
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| |
Collapse
|
8
|
Onyango S, Mi JD, Koech A, Okiro P, Temmerman M, von Dadelszen P, Tribe RM, Omuse G, the PRECISE Network. Microbiota dynamics, metabolic and immune interactions in the cervicovaginal environment and their role in spontaneous preterm birth. Front Immunol 2023; 14:1306473. [PMID: 38196946 PMCID: PMC10774218 DOI: 10.3389/fimmu.2023.1306473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024] Open
Abstract
Differences in the cervicovaginal microbiota are associated with spontaneous preterm birth (sPTB), a significant cause of infant morbidity and mortality. Although establishing a direct causal link between cervicovaginal microbiota and sPTB remains challenging, recent advancements in sequencing technologies have facilitated the identification of microbial markers potentially linked to sPTB. Despite variations in findings, a recurring observation suggests that sPTB is associated with a more diverse and less stable vaginal microbiota across pregnancy trimesters. It is hypothesized that sPTB risk is likely to be modified via an intricate host-microbe interactions rather than due to the presence of a single microbial taxon or broad community state. Nonetheless, lactobacilli dominance is generally associated with term outcomes and contributes to a healthy vaginal environment through the production of lactic acid/maintenance of a low pH that excludes other pathogenic microorganisms. Additionally, the innate immunity of the host and metabolic interactions between cervicovaginal microbiota, such as the production of bacteriocins and the use of proteolytic enzymes, exerts a profound influence on microbial populations, activities, and host immune responses. These interplays collectively impact pregnancy outcomes. This review aims to summarize the complexity of cervicovaginal environment and microbiota dynamics, and associations with bacterial vaginosis and sPTB. There is also consideration on how probiotics may mitigate the risk of sPTB and bacterial vaginosis.
Collapse
Affiliation(s)
- Stanley Onyango
- Department of Pathology, Aga Khan University, Nairobi, Kenya
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Jia Dai Mi
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Angela Koech
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Patricia Okiro
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Marleen Temmerman
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Peter von Dadelszen
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Rachel M. Tribe
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Geoffrey Omuse
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | | |
Collapse
|
9
|
Fujishiro S, Tsuji S, Akagawa S, Akagawa Y, Yamanouchi S, Ishizaki Y, Hashiyada M, Akane A, Kaneko K. Dysbiosis in Gut Microbiota in Children Born Preterm Who Developed Autism Spectrum Disorder: A Pilot Study. J Autism Dev Disord 2023; 53:4012-4020. [PMID: 35909184 DOI: 10.1007/s10803-022-05682-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 10/16/2022]
Abstract
The gut microbiota was reported to differ between children with autism spectrum disorder (ASD) and typically developing (TD) children, and dysbiosis of the gut microbiota in preterm infants is common. Here, we explored the characteristics of gut microbiota in children born preterm with ASD. We performed 16S rRNA gene sequencing using stool samples from ASD children born preterm and TD children born preterm. Alpha diversity was significantly greater in the ASD group. A comparison of beta diversity showed different clusters. Linear discriminant analysis effect size analysis revealed significantly more Firmicutes in the ASD group compared with the TD group. In conclusion, the gut microbiota in children born preterm differs between children with ASD and TD.
Collapse
Affiliation(s)
- Sadayuki Fujishiro
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Yuko Akagawa
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Yuko Ishizaki
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Masaki Hashiyada
- Department of Legal Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Atsushi Akane
- Department of Legal Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
10
|
Krupa-Kotara K, Grajek M, Grot M, Czarnota M, Wypych-Ślusarska A, Oleksiuk K, Głogowska-Ligus J, Słowiński J. Pre- and Postnatal Determinants Shaping the Microbiome of the Newborn in the Opinion of Pregnant Women from Silesia (Poland). Life (Basel) 2023; 13:1383. [PMID: 37374165 PMCID: PMC10305644 DOI: 10.3390/life13061383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Pre- and postnatal factors influence the formation of the newborn's microbiome as early as birth and the intrauterine period has a substantial impact on the composition of the baby's gastrointestinal microbiota and its subsequent development. This study intends to measure pregnant women's knowledge of the importance of microbiota for the health of the newborn. The sample was selected based on defined inclusion and exclusion criteria. The assessment of women's knowledge was assessed by the Kolmogorov-Smirnov and Kruskal-Wallis statistical tests. This study population comprised 291 adult pregnant women with a mean age of 28.4 ± 4.7 years. A total of 34% (n = 99), 35% (n = 101), and 31.3% (n = 91) were at the 1-3 trimester, respectively. The results showed that 36.4% of the women were aware that the intrauterine period changes the makeup of the gastrointestinal microbiota, whereas 5.8% exhibited awareness of the composition of the child's normal gut microbiota. Most of the women surveyed-(72.1%)-know that colonization of the tract occurs as early as the birth period. Women with student status (those who will pursue higher education in the future) and those who had given birth to the most children exhibited higher levels of knowledge.
Collapse
Affiliation(s)
- Karolina Krupa-Kotara
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Mateusz Grajek
- Department of Public Health, Department of Public Health Policy, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland;
| | - Martina Grot
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
- Doctoral School, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Martina Czarnota
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
| | - Agata Wypych-Ślusarska
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Klaudia Oleksiuk
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Joanna Głogowska-Ligus
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Jerzy Słowiński
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| |
Collapse
|
11
|
Radford-Smith DE, Anthony DC. Prebiotic and Probiotic Modulation of the Microbiota-Gut-Brain Axis in Depression. Nutrients 2023; 15:nu15081880. [PMID: 37111100 PMCID: PMC10146605 DOI: 10.3390/nu15081880] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging evidence demonstrates that alterations to the gut microbiota can affect mood, suggesting that the microbiota-gut-brain (MGB) axis contributes to the pathogenesis of depression. Many of these pathways overlap with the way in which the gut microbiota are thought to contribute to metabolic disease progression and obesity. In rodents, prebiotics and probiotics have been shown to modulate the composition and function of the gut microbiota. Together with germ-free rodent models, probiotics have provided compelling evidence for a causal relationship between microbes, microbial metabolites, and altered neurochemical signalling and inflammatory pathways in the brain. In humans, probiotic supplementation has demonstrated modest antidepressant effects in individuals with depressive symptoms, though more studies in clinically relevant populations are needed. This review critically discusses the role of the MGB axis in depression pathophysiology, integrating preclinical and clinical evidence, as well as the putative routes of communication between the microbiota-gut interface and the brain. A critical overview of the current approaches to investigating microbiome changes in depression is provided. To effectively translate preclinical breakthroughs in MGB axis research into novel therapies, rigorous placebo-controlled trials alongside a mechanistic and biochemical understanding of prebiotic and probiotic action are required from future research.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX3 7JX, UK
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
12
|
The Use of Prebiotics from Pregnancy and Its Complications: Health for Mother and Offspring—A Narrative Review. Foods 2023; 12:foods12061148. [PMID: 36981075 PMCID: PMC10048320 DOI: 10.3390/foods12061148] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Pregnancy involves a metabolic reprogramming that includes changes in the gut microbiota composition in women. Evidence shows that maternal dysbiosis is linked to neonatal dysbiosis, and this factor can determine health status in adulthood. Although there is little literature available on this topic, high heterogeneity is a limitation when examining nutritional interventions. Information has been gathered to contrast the benefits of prebiotic usage, specifically in pregnancy, in its possible complications and in newborns’ gut microbiota development. The objective pursued in this brief narrative review is to provide a clear summary of relevant content when searching with regard to the use of prebiotics in pregnancy, the effects in prenatal and postnatal periods, and to help in clinical decision-making in pregnancy management and lactation. A search has found that the nutritional status of the pregnant mother is key for the earliest microbial colonization in newborns, and thus intervention programs from pregnancy could assure better outcomes in both the mother and offspring. In this sense, prebiotics (administered to mothers who breastfeed or provided in formula milk) are feasible and cost-effective elements that can prevent allergies, colic, and other maladies in newborns.
Collapse
|
13
|
Jeong S. Factors influencing development of the infant microbiota: from prenatal period to early infancy. Clin Exp Pediatr 2022; 65:439-447. [PMID: 34942687 PMCID: PMC9441613 DOI: 10.3345/cep.2021.00955] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/03/2021] [Indexed: 11/27/2022] Open
Abstract
During early life, the gut microbial composition rapidly changes by maternal microbiota composition, delivery mode, infant feeding mode, antibiotic usage, and various environmental factors, such as the presence of pets and siblings. An integrative study on the diet, the microbiota, and genomic activity at the transcriptomic level may give an insight into the role of diet in shaping the human/microbiome relationship. Disruption in the gut microbiota (i.e., gut dysbiosis) has been linked to necrotizing enterocolitis in infancy, as well as some chronic diseases in later, including obesity, diabetes, inflammatory bowel disease, cancer, allergies, and asthma. Therefore, understanding the impact of maternal-to-infant transfer of dysbiotic microbes and then modifying infant early colonization or correcting early-life gut dysbiosis might be a potential strategy to overcome chronic health conditions.
Collapse
Affiliation(s)
- Sujin Jeong
- Division of Gastroenterology and Nutrition of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
14
|
The Gut Microbiota Dysbiosis in Preeclampsia Contributed to Trophoblast Cell Proliferation, Invasion, and Migration via lncRNA BC030099/NF-κB Pathway. Mediators Inflamm 2022; 2022:6367264. [PMID: 35784173 PMCID: PMC9249531 DOI: 10.1155/2022/6367264] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background Preeclampsia (PE) is the main reason of maternal and perinatal morbidity and mortality. Gut microbiota imbalance in PE patients is accompanied by elevated serum lipopolysaccharide (LPS) levels, but whether it affects the occurrence and development of PE, the underlying mechanism is not clear. This paper intends to investigate the relationship between lncRNA BC030099, inflammation, and gut microbiota in PE. Methods The feces of the patients were collected, and gut microbiota changes were assessed by 16S rRNA sequencing and pathway analysis by PICRUSt. Next, we examined LPS and lncRNA BC030099 levels in feces or placenta of PE patients. Then, we knocked down lncRNA BC030099 in HTR-8/SVneo cells and added the NF-κB pathway inhibitor JSH-23. CCK-8 and Transwell assays were performed to determine cell proliferation, migration, and invasion. Western blot was utilized to evaluate MMP2, MMP9, snail, and E-cadherin, p-IκBα, IκBα, and nuclear NF-κB p65 levels. IL-6, IL-1β, and TNF-α levels were examined by ELISA. Results Gut microbiota was altered in PE patients, and microbial genes associated with LPS biosynthesis were significantly elevated in gut microbiota in the PE group. LPS level in feces and placenta of PE group was significantly elevated. lncRNA BC030099 level in placenta of PE group was also notably promoted. Knockdown of lncRNA BC030099 promoted HTR-8/SVneo cell proliferation, migration, and invasion. Knockdown of lncRNA BC030099 also elevated MMP2, MMP9, and snail levels and repressed E-cadherin level. In addition, lncRNA BC030099 affected NF-κB pathway. Furthermore, NF-κB inhibitor reversed HTR-8/SVneo cell proliferation, invasion, and migration induced by LPS. Conclusions The gut microbiota dysbiosis in PE contributed to HTR-8/SVneo cell proliferation, invasion, and migration via lncRNA BC030099/NF-κB pathway.
Collapse
|
15
|
Van Daele E, Kamphorst K, Vlieger AM, Hermes G, Milani C, Ventura M, Belzer C, Smidt H, van Elburg RM, Knol J. Effect of antibiotics in the first week of life on faecal microbiota development. Arch Dis Child Fetal Neonatal Ed 2022; 107:fetalneonatal-2021-322861. [PMID: 35534183 PMCID: PMC9606546 DOI: 10.1136/archdischild-2021-322861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Infants are frequently exposed to antibiotics (AB) in the first week of life for suspected bacterial infections. Little is known about the effect of AB on the developing intestinal microbiota. Therefore, we studied intestinal microbiota development with and without AB exposure in the first week of life in term born infants. METHODS We analysed the faecal microbiota from birth until 2.5 years of age by 16S rRNA gene amplicon sequencing in a cohort with 56 term born infants, exposed to AB in the first week of life (AB+) (AB for 2-3 days (AB2, n=20), AB for 7 days (AB7, n=36)), compared with 126 healthy controls (AB-). The effects of AB and duration were examined in relation to delivery and feeding mode. RESULTS AB+ was associated with significantly increased relative abundance of Enterobacteriaceae at 3 weeks and 1 year and a decrease of Bifidobacteriaceae, from 1 week until 3 months of age only in vaginally delivered, but not in C-section born infants. Similar deviations were noted in AB7, but not in AB2. After AB, breastfed infants had lower relative abundance of potentially pathogenic Enterobacteriaceae compared with formula fed infants and recovered 2 weeks faster towards controls. CONCLUSIONS AB exposure in the first week of life alters faecal microbiota development with deviations in the relative abundance of individual taxa until 1 year of age. These alterations can have long-term health consequences, which emphasises the need for future studies aiming at restoring intestinal microbiota after AB administration.
Collapse
Affiliation(s)
- Emmy Van Daele
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Kim Kamphorst
- Pediatrics, Amsterdam Gastroenterology, Metabolism & Nutrition, Amsterdam Reproduction & Development, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Pediatrics, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Arine M Vlieger
- Pediatrics, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Gerben Hermes
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma Department of Chemical Life Sciences and Environmental Sustainability, Parma, Emilia-Romagna, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Emilia-Romagna, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma Department of Chemical Life Sciences and Environmental Sustainability, Parma, Emilia-Romagna, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Emilia-Romagna, Italy
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ruurd M van Elburg
- Pediatrics, Amsterdam Gastroenterology, Metabolism & Nutrition, Amsterdam Reproduction & Development Amsterdam, Amsterdam UMC Locatie AMC, Amsterdam, North Holland, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Nutricia Research BV, Utrecht, The Netherlands
| |
Collapse
|
16
|
Seif El Dahan K, Bejjani J, Nasrallah AA, Youssef L, Mladenovic A, Dosch L, Leone A, Jurjus A. Probiotics Properties: A Focus on Pregnancy Outcomes. Eur J Obstet Gynecol Reprod Biol 2022; 272:16-23. [PMID: 35278924 DOI: 10.1016/j.ejogrb.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/01/2022] [Accepted: 03/01/2022] [Indexed: 11/04/2022]
Abstract
A healthy microbiome plays an important role in the prevention of illness and maintenance of overall health, including reproductive health. Although the therapeutic advantages of probiotics have been shown to run across multiple organ systems, their role in pregnancy is not well explored. The aim of this review is to highlight the potential advantages and adverse effects of probiotics in pregnancy. Data were collected from the literature over the past decade using PubMed, Medline, Google Scholar, Ovid, Scopus, and Science Direct. A total of 40 articles were utilized in this review. Collected data indicated that prenatal and post-natal supplementation with lactobacilli alone or lactobacilli with Bifidobacterium spp. seems to be protective. Probiotics may improve insulin resistance and consequently reduce the risk of gestational diabetes. Probiotics may also reduce anxiety and depression by influencing brain activity. Additionally, they interfere with vaginal flora to make it friendlier to beneficial bacteria, and enhance anti-inflammatory or reduce pro-inflammatory cytokines. They may also decrease eczema in breastfed infants and prevent allergic reactions by downregulating Th2 responses to specific allergens from mid to late gestation. Leveraging the cervicovaginal microbiota could promote a number of positive pregnancy-related health outcomes. Caution should be exercised in the selection, dosing, and monitoring of probiotics administration. More comprehensive randomized clinical trials are needed to reach a more meaningful evidence-based clinical knowledge.
Collapse
Affiliation(s)
| | - Joseph Bejjani
- Faculty of Medicine, American University of Beirut, Lebanon.
| | - Ali A Nasrallah
- Faculty of Medicine, American University of Beirut, Lebanon.
| | - Lara Youssef
- Faculty of Medicine, University of Balamand, Koura, Lebanon.
| | | | - Laura Dosch
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Lebanon.
| | - Angelo Leone
- Department of Experimental Biomedicine and Neuroscience, Section of Histology and Embryology, University of Palermo, Italy.
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Lebanon.
| |
Collapse
|
17
|
Vitacolonna E, Masulli M, Palmisano L, Stuppia L, Franzago M. Inositols, Probiotics, and Gestational Diabetes: Clinical and Epigenetic Aspects. Nutrients 2022; 14:1543. [PMID: 35458105 PMCID: PMC9028601 DOI: 10.3390/nu14081543] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
There is growing interest in the potential role of different stereoisomers of inositol or their combination as well as probiotics supplementation in healthy glucose metabolism during pregnancy and in promoting offspring health. The aim of this review is to clarify the effects of several inositol and probiotics-based supplements in the prevention and treatment of gestational diabetes (GDM). Moreover, we will discuss the epigenetic aspects and their short- and long-term effects in response to probiotic intervention as well as the possible implications of these findings in guiding appropriate supplementation regimens in pregnancy.
Collapse
Affiliation(s)
- Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| | - Maria Masulli
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (M.M.); (L.P.)
| | - Luisa Palmisano
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (M.M.); (L.P.)
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy
| | - Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
18
|
Jiao L, Su CW, Cao T, Zheng S, Walker WA, Shi HN. Maternal Influences and Intervention Strategies on the Development of Food Allergy in Offspring. Front Immunol 2022; 13:817062. [PMID: 35281070 PMCID: PMC8904425 DOI: 10.3389/fimmu.2022.817062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/01/2022] [Indexed: 11/24/2022] Open
Abstract
Food allergies and other immune-mediated diseases have become serious health concerns amongst infants and children in developed and developing countries. The absence of available cures limits disease management to allergen avoidance and symptomatic treatments. Research has suggested that the presence of maternal food allergies may expose the offspring to genetic predisposition, making them more susceptible to allergen sensitization. The following review has focused on epidemiologic studies regarding maternal influences of proneness to develop food allergy in offspring. The search strategy was "food allergy OR maternal effects OR offspring OR prevention". A systematically search from PubMed/MEDLINE, Science Direct and Google Scholar was conducted. Specifically, it discussed the effects of maternal immunity, microbiota, breastfeeding, genotype and allergy exposure on the development of food allergy in offspring. In addition, several commonly utilized prenatal and postpartum strategies to reduce food allergy proneness were presented, including early diagnosis of high-risk infants and various dietary interventions.
Collapse
Affiliation(s)
- Lefei Jiao
- School of Marine Sciences, Ningbo University, Ningbo, China
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Tinglan Cao
- Laboratory for Lipid Medicine and Technology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Shasha Zheng
- Department of Nutrition, California Baptist University, Riverside, CA, United States
| | - W. Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
19
|
Aguilar-Lopez M, Dinsmoor AM, Ho TTB, Donovan SM. A systematic review of the factors influencing microbial colonization of the preterm infant gut. Gut Microbes 2022; 13:1-33. [PMID: 33818293 PMCID: PMC8023245 DOI: 10.1080/19490976.2021.1884514] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Prematurity coupled with the necessary clinical management of preterm (PT) infants introduces multiple factors that can interfere with microbial colonization. This study aimed to review the perinatal, physiological, pharmacological, dietary, and environmental factors associated with gut microbiota of PT infants. A total of 587 articles were retrieved from a search of multiple databases. Sixty studies were included in the review after removing duplicates and articles that did not meet the inclusion criteria. Review of this literature revealed that evidence converged on the effect of postnatal age, mode of delivery, use of antibiotics, and consumption of human milk in the composition of gut microbiota of PT infants. Less evidence was found for associations with race, sex, use of different fortifiers, macronutrients, and other medications. Future studies with rich metadata are needed to further explore the impact of the PT exposome on the development of the microbiota in this high-risk population.
Collapse
Affiliation(s)
- Miriam Aguilar-Lopez
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Andrew M. Dinsmoor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Thao T. B. Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, USA,CONTACT Sharon M. Donovan Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 339 Bevier Hall 905 S. Goodwin Avenue, Urbana, IL61801, USA
| |
Collapse
|
20
|
Yu D, Meng X, de Vos WM, Wu H, Fang X, Maiti AK. Implications of Gut Microbiota in Complex Human Diseases. Int J Mol Sci 2021; 22:12661. [PMID: 34884466 PMCID: PMC8657718 DOI: 10.3390/ijms222312661] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Humans, throughout the life cycle, from birth to death, are accompanied by the presence of gut microbes. Environmental factors, lifestyle, age and other factors can affect the balance of intestinal microbiota and their impact on human health. A large amount of data show that dietary, prebiotics, antibiotics can regulate various diseases through gut microbes. In this review, we focus on the role of gut microbes in the development of metabolic, gastrointestinal, neurological, immune diseases and, cancer. We also discuss the interaction between gut microbes and the host with respect to their beneficial and harmful effects, including their metabolites, microbial enzymes, small molecules and inflammatory molecules. More specifically, we evaluate the potential ability of gut microbes to cure diseases through Fecal Microbial Transplantation (FMT), which is expected to become a new type of clinical strategy for the treatment of various diseases.
Collapse
Affiliation(s)
- Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Xin Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Amit K. Maiti
- Department of Genetics and Genomics, Mydnavar, 2645 Somerset Boulevard, Troy, MI 48084, USA
| |
Collapse
|
21
|
Preterm infant meconium microbiota transplant induces growth failure, inflammatory activation, and metabolic disturbances in germ-free mice. Cell Rep Med 2021; 2:100447. [PMID: 34841294 PMCID: PMC8606908 DOI: 10.1016/j.xcrm.2021.100447] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/17/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022]
Abstract
Preterm birth may result in adverse health outcomes. Very preterm infants typically exhibit postnatal growth restriction, metabolic disturbances, and exaggerated inflammatory responses. We investigated the differences in the meconium microbiota composition between very preterm (<32 weeks), moderately preterm (32–37 weeks), and term (>37 weeks) human neonates by 16S rRNA gene sequencing. Human meconium microbiota transplants to germ-free mice were conducted to investigate whether the meconium microbiota is causally related to the preterm infant phenotype in an experimental model. Our results indicate that very preterm birth is associated with a distinct meconium microbiota composition. Fecal microbiota transplant of very preterm infant meconium results in impaired growth, altered intestinal immune function, and metabolic parameters as compared to term infant meconium transplants in germ-free mice. This finding suggests that measures aiming to minimize the long-term adverse consequences of very preterm birth should be commenced during pregnancy or directly after birth. Very preterm neonates exhibit a distinct meconium microbiota composition Human meconium microbiota is transplanted to germ-free mice in this study Preterm transplant induces growth restriction, inflammation, and altered metabolism Initial gut microbiota may be causally related to complications of prematurity
Collapse
|
22
|
Selma-Royo M, Calvo Lerma J, Cortés-Macías E, Collado MC. Human milk microbiome: From actual knowledge to future perspective. Semin Perinatol 2021; 45:151450. [PMID: 34274151 DOI: 10.1016/j.semperi.2021.151450] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is the gold standard for infant nutrition during the first months of life since it is perfectly adapted to the neonatal nutritional requirements and supports infant growth and development. Human milk contains a complex nutritional and bioactive composition including microorganisms and oligosaccharides which would also contribute to the gut and immune system maturation. Despite the growing evidence, the factors contributing to milk microbes' variations and the potential functions on the infant's gut are still uncovered. This short-review provides a general overview of milk microbiota, potential factors shaping its composition, contribution to the infant microbiota and immune system development, including the suggested biological relevance for infant health as well as the description of tools and strategies aimed to restore and module microbes in milk.
Collapse
Affiliation(s)
- Marta Selma-Royo
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| | - Joaquim Calvo Lerma
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Erika Cortés-Macías
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| |
Collapse
|
23
|
Ansari A, Bose S, You Y, Park S, Kim Y. Molecular Mechanism of Microbiota Metabolites in Preterm Birth: Pathological and Therapeutic Insights. Int J Mol Sci 2021; 22:8145. [PMID: 34360908 PMCID: PMC8347546 DOI: 10.3390/ijms22158145] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Preterm birth (PTB) refers to the birth of infants before 37 weeks of gestation and is a challenging issue worldwide. Evidence reveals that PTB is a multifactorial dysregulation mediated by a complex molecular mechanism. Thus, a better understanding of the complex molecular mechanisms underlying PTB is a prerequisite to explore effective therapeutic approaches. During early pregnancy, various physiological and metabolic changes occur as a result of endocrine and immune metabolism. The microbiota controls the physiological and metabolic mechanism of the host homeostasis, and dysbiosis of maternal microbial homeostasis dysregulates the mechanistic of fetal developmental processes and directly affects the birth outcome. Accumulating evidence indicates that metabolic dysregulation in the maternal or fetal membranes stimulates the inflammatory cytokines, which may positively progress the PTB. Although labour is regarded as an inflammatory process, it is still unclear how microbial dysbiosis could regulate the molecular mechanism of PTB. In this review based on recent research, we focused on both the pathological and therapeutic contribution of microbiota-generated metabolites to PTB and the possible molecular mechanisms.
Collapse
Affiliation(s)
- AbuZar Ansari
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Shambhunath Bose
- Department of Bioscience, Sri Sathya Sai University for Human Excellence, Navanihal, Okali Post, Kamalapur, Kalaburagi, Karnataka 585313, India;
| | - Youngah You
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Sunwha Park
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Youngju Kim
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| |
Collapse
|
24
|
Miao T, Yu Y, Sun J, Ma A, Yu J, Cui M, Yang L, Wang H. Decrease in abundance of bacteria of the genus Bifidobacterium in gut microbiota may be related to pre-eclampsia progression in women from East China. Food Nutr Res 2021; 65:5781. [PMID: 34262418 PMCID: PMC8254465 DOI: 10.29219/fnr.v65.5781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/15/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pre-eclampsia (PE) can result in severe damage to maternal and fetal health. It has been reported that gut microbiota (GM) had important roles in regulating the metabolic and inflammatory responses of the mother. However, investigations on GM in PE are rare. Objective The objective of the present study was to investigate the changes of GM in PE and how to alter the GM composition in PE by dietary or dietary supplements. Design We analyzed the composition changes in GM as well as the relationship between bacteria of different genera and clinical indices by amplifying the V4 region of the 16S ribosomal RNA gene in 12 PE patients and eight healthy pregnant women in East China. Results In the PE group, the Observed Species Index was lower than that in the control group, indicating that the α-diversity of the microbiome in the PE group decreased. At phylum, family, and genus levels, the relative abundance of different bacteria in PE patients displayed substantial differences to those from healthy women. We noted a decreased abundance of bacteria of the phylum Actinobacteria (P = 0.042), decreased abundance of bacteria of the family Bifidobacteriaceae (P = 0.039), increased abundance of bacteria of the genus Blautia (P = 0.026) and Ruminococcus (P = 0.048), and decreased abundance of bacteria of the genus Bifidobacterium (P = 0.038). Among three enriched genera, bacteria of the genus Bifidobacterium showed a negative correlation with the systolic blood pressure (SBP), diastolic blood pressure (DBP), and dyslipidemia, which involved glucose metabolism, lipid metabolism, and the oxidative-phosphorylation pathway. The increased abundance of bacteria of the genera Blautia and Ruminococcus was positively correlated with obesity and dyslipidemia, which involved lipid metabolism, glycosyltransferases, biotin metabolism, and the oxidative-phosphorylation pathways. Moreover, women in the PE group ate more than women in the control group, so fetuses were more prone to overnutrition in the PE group. Conclusion There is a potential for GM dysbiosis in PE patients, and they could be prone to suffer from metabolic syndrome. We speculate that alterations in the abundance of bacteria of certain genera (e.g. increased abundance of Blautia and Ruminococcus, and decreased abundance of Bifidobacterium) were associated with PE development to some degree. Our data could help to monitor the health of pregnant women and may be helpful for preventing and assisting treatment of PE by increasing dietary fiber or probiotics supplement.
Collapse
Affiliation(s)
- Tingting Miao
- Department of Education, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China.,Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Yun Yu
- Department of Clinical Laboratory, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Aiguo Ma
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jinran Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Mengjun Cui
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Liping Yang
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Huiyan Wang
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
25
|
Mukherjee S, Mitra S, Dutta S, Basu S. Neonatal Sepsis: The Impact of Carbapenem-Resistant and Hypervirulent Klebsiella pneumoniae. Front Med (Lausanne) 2021; 8:634349. [PMID: 34179032 PMCID: PMC8225938 DOI: 10.3389/fmed.2021.634349] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/26/2021] [Indexed: 01/12/2023] Open
Abstract
The convergence of a vulnerable population and a notorious pathogen is devastating, as seen in the case of sepsis occurring during the first 28 days of life (neonatal period). Sepsis leads to mortality, particularly in low-income countries (LICs) and lower-middle-income countries (LMICs). Klebsiella pneumoniae, an opportunistic pathogen is a leading cause of neonatal sepsis. The success of K. pneumoniae as a pathogen can be attributed to its multidrug-resistance and hypervirulent-pathotype. Though the WHO still recommends ampicillin and gentamicin for the treatment of neonatal sepsis, K. pneumoniae is rapidly becoming untreatable in this susceptible population. With escalating rates of cephalosporin use in health-care settings, the increasing dependency on carbapenems, a "last resort antibiotic," has led to the emergence of carbapenem-resistant K. pneumoniae (CRKP). CRKP is reported from around the world causing outbreaks of neonatal infections. Carbapenem resistance in CRKP is largely mediated by highly transmissible plasmid-encoded carbapenemase enzymes, including KPC, NDM, and OXA-48-like enzymes. Further, the emergence of a more invasive and highly pathogenic hypervirulent K. pneumoniae (hvKP) pathotype in the clinical context poses an additional challenge to the clinicians. The deadly package of resistance and virulence has already limited therapeutic options in neonates with a compromised defense system. Although there are reports of CRKP infections, a review on neonatal sepsis due to CRKP/ hvKP is scarce. Here, we discuss the current understanding of neonatal sepsis with a focus on the global impact of the CRKP, provide a perspective regarding the possible acquisition and transmission of the CRKP and/or hvKP in neonates, and present strategies to effectively identify and combat these organisms.
Collapse
Affiliation(s)
- Subhankar Mukherjee
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shravani Mitra
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sulagna Basu
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
26
|
Mate A, Reyes-Goya C, Santana-Garrido Á, Vázquez CM. Lifestyle, Maternal Nutrition and Healthy Pregnancy. Curr Vasc Pharmacol 2021; 19:132-140. [PMID: 32234002 DOI: 10.2174/1570161118666200401112955] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 02/07/2023]
Abstract
Healthy lifestyle habits spanning from preconception to postpartum are considered as a major safeguard for achieving successful pregnancies and for the prevention of gestational diseases. Among preconception priorities established by the World Health Organization (WHO) are healthy diet and nutrition, weight management, physical activity, planned pregnancy and physical, mental and psychosocial health. Most studies covering the topic of healthy pregnancies focus on maternal diet because obesity increases the risks for adverse perinatal outcomes, including gestational diabetes mellitus, large for gestational age newborns, or preeclampsia. Thus, foods rich in vegetables, essential and polyunsaturated fats and fibre-rich carbohydrates should be promoted especially in overweight, obese or diabetic women. An adequate intake of micronutrients (e.g. iron, calcium, folate, vitamin D and carotenoids) is also crucial to support pregnancy and breastfeeding. Moderate physical activity throughout pregnancy improves muscle tone and function, besides decreasing the risk of preeclampsia, gestational diabesity (i.e. diabetes associated with obesity) and postpartum overweight. Intervention studies claim that an average of 30 min of exercise/day contributes to long-term benefits for maternal overall health and wellbeing. Other factors such as microbiome modulation, behavioural strategies (e.g. smoking cessation, anxiety/stress reduction and sleep quality), maternal genetics and age, social class and education might also influence the maternal quality of life. These factors contribute to ensure a healthy pregnancy, or at least to reduce the risk of adverse maternal and foetal outcomes during pregnancy and later in life.
Collapse
Affiliation(s)
- Alfonso Mate
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain
| | - Claudia Reyes-Goya
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain
| | - Álvaro Santana-Garrido
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain
| | - Carmen M Vázquez
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain
| |
Collapse
|
27
|
Nunez N, Réot L, Menu E. Neonatal Immune System Ontogeny: The Role of Maternal Microbiota and Associated Factors. How Might the Non-Human Primate Model Enlighten the Path? Vaccines (Basel) 2021; 9:584. [PMID: 34206053 PMCID: PMC8230289 DOI: 10.3390/vaccines9060584] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/23/2022] Open
Abstract
Interactions between the immune system and the microbiome play a crucial role on the human health. These interactions start in the prenatal period and are critical for the maturation of the immune system in newborns and infants. Several factors influence the composition of the infant's microbiota and subsequently the development of the immune system. They include maternal infection, antibiotic treatment, environmental exposure, mode of delivery, breastfeeding, and food introduction. In this review, we focus on the ontogeny of the immune system and its association to microbial colonization from conception to food diversification. In this context, we give an overview of the mother-fetus interactions during pregnancy, the impact of the time of birth and the mode of delivery, the neonate gastrointestinal colonization and the role of breastfeeding, weaning, and food diversification. We further review the impact of the vaccination on the infant's microbiota and the reciprocal case. Finally, we discuss several potential therapeutic interventions that might help to improve the newborn and infant's health and their responses to vaccination. Throughout the review, we underline the main scientific questions that are left to be answered and how the non-human primate model could help enlighten the path.
Collapse
Affiliation(s)
- Natalia Nunez
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
| | - Louis Réot
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
| | - Elisabeth Menu
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
- MISTIC Group, Department of Virology, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
28
|
Ishimwe JA. Maternal microbiome in preeclampsia pathophysiology and implications on offspring health. Physiol Rep 2021; 9:e14875. [PMID: 34042284 PMCID: PMC8157769 DOI: 10.14814/phy2.14875] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
Preeclampsia is a devastating hypertensive pregnancy disorder that currently affects 2%–8% of pregnancies worldwide. It is associated with maternal and fetal mortality and morbidity and adverse health outcomes both in mom and offspring beyond pregnancy. The pathophysiology is not completely understood, and there are no approved therapies to specifically treat for the disease, with only few therapies approved to manage symptoms. Recent advances suggest that aberrations in the composition of the microbiome may play a role in the pathogenesis of various diseases including preeclampsia. The maternal and uteroplacental environments greatly influence the long‐term health outcomes of the offspring through developmental programming mechanisms. The current review summarizes recent developments on the role of the microbiome in adverse pregnancy outcomes with a focus on preeclampsia. It also discusses the potential role of the maternal microbiome in fetal programming; explores gut‐targeted therapeutics advancement and their implications in the treatment of preeclampsia.
Collapse
Affiliation(s)
- Jeanne A Ishimwe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
29
|
Chleilat F, Schick A, Reimer RA. Microbiota Changes in Fathers Consuming a High Prebiotic Fiber Diet Have Minimal Effects on Male and Female Offspring in Rats. Nutrients 2021; 13:820. [PMID: 33801321 PMCID: PMC8001975 DOI: 10.3390/nu13030820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Consuming a diet high in prebiotic fiber has been associated with improved metabolic and gut microbial parameters intergenerationally, although studies have been limited to maternal intake with no studies examining this effect in a paternal model. METHOD Male Sprague Dawley rats were allocated to either (1) control or (2) oligofructose-supplemented diet for nine weeks and then mated. Offspring consumed control diet until 16 weeks of age. Bodyweight, body composition, glycemia, hepatic triglycerides, gastrointestinal hormones, and gut microbiota composition were measured in fathers and offspring. RESULTS Paternal energy intake was reduced, while satiety inducing peptide tyrosine tyrosine (PYY) gut hormone was increased in prebiotic versus control fathers. Increased serum PYY persisted in female prebiotic adult offspring. Hepatic triglycerides were decreased in prebiotic fathers with a similar trend (p = 0.07) seen in female offspring. Gut microbial composition showed significantly reduced alpha diversity in prebiotic fathers at 9 and 12 weeks of age (p < 0.001), as well as concurrent differences in beta diversity (p < 0.001), characterized by differences in Bifidobacteriaceae, Lactobacillaceae and Erysipelotrichaceae, and particularly Bifidobacterium animalis. Female prebiotic offspring had higher alpha diversity at 3 and 9 weeks of age (p < 0.002) and differences in beta diversity at 15 weeks of age (p = 0.04). Increases in Bacteroidetes in female offspring and Christensenellaceae in male offspring were seen at nine weeks of age. CONCLUSIONS Although paternal prebiotic intake before conception improves metabolic and microbiota outcomes in fathers, effects on offspring were limited with increased serum satiety hormone levels and changes to only select gut bacteria.
Collapse
Affiliation(s)
- Faye Chleilat
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Raylene A. Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
30
|
Kurkjian HM, Akbari MJ, Momeni B. The impact of interactions on invasion and colonization resistance in microbial communities. PLoS Comput Biol 2021; 17:e1008643. [PMID: 33481772 PMCID: PMC7857599 DOI: 10.1371/journal.pcbi.1008643] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/03/2021] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
In human microbiota, the prevention or promotion of invasions can be crucial to human health. Invasion outcomes, in turn, are impacted by the composition of resident communities and interactions of resident members with the invader. Here we study how interactions influence invasion outcomes in microbial communities, when interactions are primarily mediated by chemicals that are released into or consumed from the environment. We use a previously developed dynamic model which explicitly includes species abundances and the concentrations of chemicals that mediate species interaction. Using this model, we assessed how species interactions impact invasion by simulating a new species being introduced into an existing resident community. We classified invasion outcomes as resistance, augmentation, displacement, or disruption depending on whether the richness of the resident community was maintained or decreased and whether the invader was maintained in the community or went extinct. We found that as the number of invaders introduced into the resident community increased, disruption rather than augmentation became more prevalent. With more facilitation of the invader by the resident community, resistance outcomes were replaced by displacement and augmentation. By contrast, with more facilitation among residents, displacement outcomes shifted to resistance. When facilitation of the resident community by the invader was eliminated, the majority of augmentation outcomes turned into displacement, while when inhibition of residents by invaders was eliminated, invasion outcomes were largely unaffected. Our results suggest that a better understanding of interactions within resident communities and between residents and invaders is crucial to predicting the success of invasions into microbial communities. Our resident microbiota can prevent diseases by making it harder for pathogens to grow and establish, a phenomenon called “colonization resistance.” Colonization resistance is one of the major benefits provided by human-associated microbiota and a viable alternative to the use of antibiotics for preventing or treating infections. Here we use a model of microbial interactions through production and consumption of metabolic compounds to assay invasion and colonization resistance. We systematically examine in simulations how interactions among resident members and those between residents and an invader impact colonization resistance and invasion outcomes. In our simulations, the common strategy of increasing the dosage of probiotics is often unsuccessful for augmenting a new species into a resident microbiota. Instead, we find that the net facilitation or inhibition between the resident members and the invader explains whether the community remains intact and whether the invader can establish. Our results suggest that a better understanding of microbial interactions can inform successful microbiota interventions.
Collapse
Affiliation(s)
- Helen M. Kurkjian
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - M. Javad Akbari
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Babak Momeni
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Chen K, Yuan T. The role of microbiota in neonatal hyperbilirubinemia. Am J Transl Res 2020; 12:7459-7474. [PMID: 33312382 PMCID: PMC7724329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/24/2020] [Indexed: 06/12/2023]
Abstract
Hyperbilirubinemia accounts for about 60% of full-term and 80% of preterm neonates globally, which is characterized by physiologically elevated unconjugated bilirubin in serum, but abnormally high levels of bilirubin have potential neurotoxic effects. Several factors contribute to the development of neonatal hyperbilirubinemia, including isoimmunization, dysregulated gut flora, genetic alteration and environmental factors. Animal studies have pinpointed the causal roles of several bacteria in bilirubin metabolism. Human studies have revealed microbiota composition in hyperbilirubinemia and found that gut microbiota affect newborns with different severity of hyperbilirubinemia. However, dysbiosis and subsequent changes in microbiota-related metabolic processes are not always considered. This review aims to describe the critical microbiota signatures for neonatal hyperbilirubinemia and focus on the underlying pathogenetic mechanism. These scientific bases give a new and accurate therapeutic strategy for the application of gut microbiota.
Collapse
Affiliation(s)
- Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| |
Collapse
|
32
|
Pourmirzaiee MA, Famouri F, Moazeni W, Hassanzadeh A, Hajihashemi M. The efficacy of the prenatal administration of Lactobacillus reuteri LR92 DSM 26866 on the prevention of infantile colic: a randomized control trial. Eur J Pediatr 2020; 179:1619-1626. [PMID: 32372186 DOI: 10.1007/s00431-020-03641-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 11/30/2022]
Abstract
Infantile colic is a functional gastrointestinal disease of the infancy that its cause has not yet been properly identified. It leads to severe discomfort in the infants and anxiety in their mothers. Probiotics have recently been recommended as an effective treatment for the improvement of infantile colic. The objective of this study is to determine the role of prenatal administration of Lactobacillus reuteri (L. reuteri) LR92 DSM 26866 on the occurrence of infantile colic. This double-blinded, placebo-controlled, randomized trial was conducted with healthy pregnant women from December 2017 to December 2018 in Isfahan, Iran. A total of 145 patients was included in this study. The case group consisted of 87 pregnant women, who received daily doses of 1 × 108 colony-forming units of live L. reuteri LR92 DSM 26866, and the control group with 88 pregnant women received the placebo (containing 9% glucose solutions) for the last 4 weeks of pregnancy. Mothers and their infants in both groups did not have significant differences in anthropometric indices, and the infants' feeding pattern. Infants born to both groups of mothers followed for 5 months on signs and symptoms of colic with the repetitive examination by a blinded pediatrics assistant to record the occurrence of colic and its grading. Mothers who received placebo were 2.36 times more likely to have infants exhibiting infantile colic than mothers in the L. reuteri LR92 DSM 26866 group (CI 95%, 1.18-4.73). Using Mann-Whitney U test, the Mean (SD) of colic severity was significantly lower in the intervention group (p = 0.01). The frequency of colic and its higher grades were significantly lower in the intervention group (p = 0.03 for the presence of colic and p = 0.01 for high grades of colic). The frequency of colic presence and its different grades according to mothers' delivery mode and infant feeding patterns were not different between the two groups (p > 0.05).Conclusion: Maternal prenatal supplementation with probiotic L. reuteri LR92 DSM 26866 during the last 4 weeks of pregnancy can prevent the occurrence and reduce the severity of infantile colic. What is Known • Lactobacillus reuteri LR92 DSM 26866 is effective in improving the symptoms of infantile colic. What is New • Prenatal administration of Lactobacillus Reuteri LR92 DSM 26866 can prevent the occurrence of infantile colic or reduce its severity.
Collapse
Affiliation(s)
- Mohammad Ali Pourmirzaiee
- Department of Pediatrics, Imam Hossein Hospital, Isfahan, Iran.,Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan, Iran.,Department of Pediatrics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Famouri
- Department of Pediatrics, Imam Hossein Hospital, Isfahan, Iran. .,Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan, Iran. .,Department of Pediatrics, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Wida Moazeni
- Department of Pediatrics, Imam Hossein Hospital, Isfahan, Iran.,Department of Pediatrics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akbar Hassanzadeh
- Department of Statistical Sciences, Isfahan University of Medical Science, Isfahan, Iran
| | - Maryam Hajihashemi
- Department of Obstetrics and Gynecology, Beheshti Hospital, Isfahan, Iran
| |
Collapse
|
33
|
Lajqi T, Pöschl J, Frommhold D, Hudalla H. The Role of Microbiota in Neutrophil Regulation and Adaptation in Newborns. Front Immunol 2020; 11:568685. [PMID: 33133082 PMCID: PMC7550463 DOI: 10.3389/fimmu.2020.568685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Newborns are highly susceptible to infections and mainly rely on innate immune functions. Reduced reactivity, delayed activation and subsequent failure to resolve inflammation however makes the neonatal immune system a very volatile line of defense. Perinatal microbiota, nutrition and different extra-uterine factors are critical elements that define long-term outcomes and shape the immune system during the neonatal period. Neutrophils are first responders and represent a vital component of the immune system in newborns. They have long been regarded as merely executive immune cells, however this notion is beginning to shift. Neutrophils are shaped by their surrounding and adaptive elements have been described. The role of “innate immune memory” and the main triangle connection microbiome—neutrophil—adaptation will be discussed in this review.
Collapse
Affiliation(s)
- Trim Lajqi
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Johannes Pöschl
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, Memmingen, Germany
| | - Hannes Hudalla
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| |
Collapse
|
34
|
Type 2 Diabetes Mellitus Associated with Obesity (Diabesity). The Central Role of Gut Microbiota and Its Translational Applications. Nutrients 2020; 12:nu12092749. [PMID: 32917030 PMCID: PMC7551493 DOI: 10.3390/nu12092749] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is a condition of rising prevalence worldwide, with important socioeconomic implications, being considered as a growing public health concern. Frequently, obesity brings other complications in addition to itself—like Type 2 Diabetes Mellitus (T2DM)—sharing origin, risk factors and pathophysiological mechanisms. In this context, some authors have decided to include both conditions as a unique entity known as “diabesity”. In fact, understanding diabesity as a single disease is possible to maximise the benefits from therapies received in these patients. Gut microbiota plays a key role in individual’s health, and their alterations, either in its composition or derived products are related to a wide range of metabolic disorders like T2DM and obesity. The present work aims to collect the different changes reported in gut microbiota in patients with T2DM associated with obesity and their possible role in the onset, development, and establishment of the disease. Moreover, current research lines to modulate gut microbiota and the potential clinical translation derived from the knowledge of this system will also be reviewed, which may provide support for a better clinical management of such a complex condition.
Collapse
|
35
|
Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020; 8:573. [PMID: 32326636 PMCID: PMC7232163 DOI: 10.3390/microorganisms8040573] [Citation(s) in RCA: 989] [Impact Index Per Article: 197.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
The complex polymicrobial composition of human gut microbiota plays a key role in health and disease. Lachnospiraceae belong to the core of gut microbiota, colonizing the intestinal lumen from birth and increasing, in terms of species richness and their relative abundances during the host's life. Although, members of Lachnospiraceae are among the main producers of short-chain fatty acids, different taxa of Lachnospiraceae are also associated with different intra- and extraintestinal diseases. Their impact on the host physiology is often inconsistent across different studies. Here, we discuss changes in Lachnospiraceae abundances according to health and disease. With the aim of harnessing Lachnospiraceae to promote human health, we also analyze how nutrients from the host diet can influence their growth and how their metabolites can, in turn, influence host physiology.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| |
Collapse
|
36
|
Carbone F, Montecucco F, Sahebkar A. Current and emerging treatments for neonatal sepsis. Expert Opin Pharmacother 2020; 21:549-556. [PMID: 32011188 DOI: 10.1080/14656566.2020.1721464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/22/2020] [Indexed: 01/27/2023]
Abstract
Introduction: Mortality due to sepsis is still prevalent, peaking at extreme ages of life including infancy. Despite many efforts, the peculiarity of the infant immune system has limited further advances in its treatment. Indeed, neonates experience a dramatic physiological transition from immune tolerance to the maternal antigens to functional maturity. Such a transition is extremely dynamic, as is the pathophysiology of infant sepsis, which is dependent on many infant, maternal, and environmental factors.Areas covered: In this review, the authors critically update and summarize the current paradigm of immunomodulation in infant sepsis. They confirm how exogenous stimulation of the immune system through intravenous immunoglobulin, colony stimulating factors, and granulocyte transfusion have failed to impact on the prognosis of infant sepsis. They also strongly support the beneficial effects of supplementation/replacement therapies with products naturally contained within maternal milk as well as antioxidant compounds.Expert opinion: Breastfeeding is beneficial against sepsis. Knowledge of the neonatal immune system is indeed too limited to effectively strengthen immune response by exogenous interventions, especially in preterm and low-birth-weight infants. Awareness of this limitation should pave the way for future studies (e.g. gender- and omics-based) aimed at better characterizing the infant immune system and promoting a more tailored approach.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
37
|
Lactobacillus fermentum CECT5716 Supplementation in Rats during Pregnancy and Lactation Impacts Maternal and Offspring Lipid Profile, Immune System and Microbiota. Cells 2020; 9:cells9030575. [PMID: 32121244 PMCID: PMC7140451 DOI: 10.3390/cells9030575] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Probiotics have shown potential for their use in early life. This study aimed to investigate whether the administration of Lactobacillus fermentum CECT5716 during pregnancy and lactation periods impacts maternal and offspring plasma lipid profile, immune system and microbiota. Rats were supplemented with the probiotic during gestation and two weeks of lactation. After supplementation, although the microbiota composition was not affected, the probiotic strain was detected in all cecal contents of dams and in some of their pups. Dams showed reduced proportion of T cytotoxic cells in the mesenteric lymph nodes, modulation of intestinal cytokines (IL-10 and IL-12) and changes in plasma fatty acids (20:0, 22:0, 20:5 n-3, and 18:3 n-6). Pups showed changes in immunoglobulins (intestinal IgA and plasmatic IgG2a and IgG2c) and fatty acid profile (17:0, 22:0, and 18:2 n-6). Overall, Lactobacillus fermentum CECT5716 supplementation contributed to beneficially modulating the immune system of the mother and its offspring.
Collapse
|
38
|
Golofast B, Vales K. The connection between microbiome and schizophrenia. Neurosci Biobehav Rev 2019; 108:712-731. [PMID: 31821833 DOI: 10.1016/j.neubiorev.2019.12.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/01/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022]
Abstract
There has been an accumulation of knowledge about the human microbiome, some detailed investigations of the gastrointestinal microbiota and its functions, and the highlighting of complex interactions between the gut, the gut microbiota, and the central nervous system. That assumes the involvement of the microbiome in the pathogenesis of various CNS diseases, including schizophrenia. Given this information and the fact, that the gut microbiota is sensitive to internal and environmental influences, we have speculated that among the factors that influence the formation and composition of gut microbiota during life, possible key elements in the schizophrenia development chain are hidden where gut microbiota is a linking component. This article aims to describe and understand the developmental relationships between intestinal microbiota and the risk of developing schizophrenia.
Collapse
Affiliation(s)
- Bogdana Golofast
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Prague East, Czech Republic; Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic.
| | - Karel Vales
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Prague East, Czech Republic
| |
Collapse
|
39
|
Louis-Jean S, Martirosyan D. Nutritionally Attenuating the Human Gut Microbiome To Prevent and Manage Metabolic Syndrome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12675-12684. [PMID: 31661963 DOI: 10.1021/acs.jafc.9b04879] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Metabolic syndrome (MSyn) constitutes a litany of pathophysiological conditions, such as central adiposity, hypertension, dyslipidemia, and hyperglycemia. As a result of the epidemic levels of MSyn, several efforts have been made to identify the etiologies of the condition and develop methods by which to reduce its prevalence. The attenuation of the gut microflora ratio of Firmicutes/Bacteroidetes through bioactive compounds found in the Mediterranean diet, dietary polysaccharides, and pre- and probiotics can be used as functional foods to improve derangements in cardiometabolic markers correlated with the development of MSyn. Although more studies are needed to understand the role of manipulating the gut microbiota in health and disease in human models, this review based on current data from epidemiologic studies and clinical trials will serve as a review to elucidate the role nutrition plays in attenuating the gut microbiota in preventing and managing MSyn.
Collapse
Affiliation(s)
- Scarlet Louis-Jean
- Functional Food Center , Functional Food Institute , Dallas , Texas 75254 , United States
| | - Danik Martirosyan
- Functional Food Center , Functional Food Institute , Dallas , Texas 75254 , United States
| |
Collapse
|
40
|
Wang Q, Sun Q, Qi R, Wang J, Qiu X, Liu Z, Huang J. Effects of Lactobacillus plantarum on the intestinal morphology, intestinal barrier function and microbiota composition of suckling piglets. J Anim Physiol Anim Nutr (Berl) 2019; 103:1908-1918. [PMID: 31498508 DOI: 10.1111/jpn.13198] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 08/03/2019] [Indexed: 02/06/2023]
Abstract
This study investigated the effect of Lactobacillus plantarum strain 299v on gut health in suckling piglets. Sixty newborn piglets were assigned to control and probiotic treatments, with three litters per treatment (ten piglets/litter). From days 1 to 20 of life, piglets were orally administered a placebo of 0.1% peptone or 1.0 × 1010 CFU L. plantarum 299v daily. Six piglets per treatment were sacrificed on day 20, and intestinal tissues (including duodenum, jejunum, ileum and colon) and the intestinal contents from colon segments were collected. The results demonstrated that piglets treated with L. plantarum 299v had a lower diarrhoea incidence than the controls. L. plantarum 299v administration significantly increased the ratio of the villus height to the crypt depth in the jejunum and ileum, as well as the mRNA expression of jejunal occludin and ileal zonula occludens 1 (ZO-1). The L. plantarum treatment also increased the mRNA abundance of porcine β-defensin 2 (pBD2) and pBD3 in the jejunum and ileum and of toll-like receptors (TLRs), such as TLR2, TLR4, TLR6 and TLR9 in the ileum, and significantly upregulated the mRNA abundances of ileal pBD1 and colonic TLR4. Additionally, the L. plantarum 299v treatment significantly changed the structure of the colonic microbiota, as evidenced by the obvious increases in the relative abundances of the phyla Firmicutes and Actinobacteria and of the genus Lactobacillus. Our findings indicate that L. plantarum 299v facilitates the gut health of suckling piglets, probably by improving the intestinal morphology and intestinal barrier function and by modifying the structure of the gut microbiota.
Collapse
Affiliation(s)
- Qi Wang
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Qian Sun
- Chongqing Academy of Animal Sciences, Chongqing, China.,College of Animal Science, Southwest University, Chongqing, China
| | - Renli Qi
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Jing Wang
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Zuohua Liu
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| |
Collapse
|
41
|
Mills S, Lane JA, Smith GJ, Grimaldi KA, Ross RP, Stanton C. Precision Nutrition and the Microbiome Part II: Potential Opportunities and Pathways to Commercialisation. Nutrients 2019; 11:E1468. [PMID: 31252674 PMCID: PMC6683087 DOI: 10.3390/nu11071468] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Modulation of the human gut microbiota through probiotics, prebiotics and dietary fibre are recognised strategies to improve health and prevent disease. Yet we are only beginning to understand the impact of these interventions on the gut microbiota and the physiological consequences for the human host, thus forging the way towards evidence-based scientific validation. However, in many studies a percentage of participants can be defined as 'non-responders' and scientists are beginning to unravel what differentiates these from 'responders;' and it is now clear that an individual's baseline microbiota can influence an individual's response. Thus, microbiome composition can potentially serve as a biomarker to predict responsiveness to interventions, diets and dietary components enabling greater opportunities for its use towards disease prevention and health promotion. In Part I of this two-part review, we reviewed the current state of the science in terms of the gut microbiota and the role of diet and dietary components in shaping it and subsequent consequences for human health. In Part II, we examine the efficacy of gut-microbiota modulating therapies at different life stages and their potential to aid in the management of undernutrition and overnutrition. Given the significance of an individual's gut microbiota, we investigate the feasibility of microbiome testing and we discuss guidelines for evaluating the scientific validity of evidence for providing personalised microbiome-based dietary advice. Overall, this review highlights the potential value of the microbiome to prevent disease and maintain or promote health and in doing so, paves the pathway towards commercialisation.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Jonathan A Lane
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - Graeme J Smith
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | | | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Fermoy P61 C996, Co Cork, Ireland.
| |
Collapse
|
42
|
Lerner A, Shoenfeld Y, Matthias T. Probiotics: If It Does Not Help It Does Not Do Any Harm. Really? Microorganisms 2019; 7:104. [PMID: 30979072 PMCID: PMC6517882 DOI: 10.3390/microorganisms7040104] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 12/19/2022] Open
Abstract
Probiotics per definition should have beneficial effects on human health, and their consumption has tremendously increased in the last decades. In parallel, the amount of published material and claims for their beneficial efficacy soared continuously. Recently, multiple systemic reviews, meta-analyses, and expert opinions expressed criticism on their claimed effects and safety. The present review describes the dark side of the probiotics, in terms of problematic research design, incomplete reporting, lack of transparency, and under-reported safety. Highlighted are the potential virulent factors and the mode of action in the intestinal lumen, risking the physiological microbiome equilibrium. Finally, regulatory topics are discussed to lighten the heterogeneous guidelines applied worldwide. The shift in the scientific world towards a better understanding of the human microbiome, before consumption of the probiotic cargo, is highly endorsed. It is hoped that better knowledge will extend the probiotic repertoire, re-confirm efficacy or safety, establish their efficacy and substantiate their beneficial effects.
Collapse
Affiliation(s)
- Aaron Lerner
- B. Rappaport School of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel.
- AESKU.KIPP Institute, 55234 Wendelsheim, Germany.
| | - Yehuda Shoenfeld
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 5262000, Israel.
| | | |
Collapse
|
43
|
Paul HA, Collins KH, Nicolucci AC, Urbanski SJ, Hart DA, Vogel HJ, Reimer RA. Maternal prebiotic supplementation reduces fatty liver development in offspring through altered microbial and metabolomic profiles in rats. FASEB J 2019; 33:5153-5167. [PMID: 30629464 DOI: 10.1096/fj.201801551r] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A maternal high-fat/sucrose diet, in the presence of maternal obesity, can program increased susceptibility to obesity and metabolic disease in offspring. In particular, nonalcoholic fatty liver disease risk is associated with poor maternal nutrition and obesity status, which may manifest via alterations in gut microbiota. Here, we report that in a preclinical model of diet-induced maternal obesity, maternal supplementation of a high-fat/sucrose diet with the prebiotic oligofructose improves glucose tolerance, insulin sensitivity, and hepatic steatosis in offspring following a long-term high-fat/sucrose dietary challenge compared with offspring of untreated dams. These improvements are associated with alterations in gut microbial composition and serum inflammatory profiles in early life and improvements in inflammatory and fatty-acid gene expression profiles in tissues. Serum metabolomics analysis highlights potential metabolic links between the gut microbiota and the degree of steatosis, including alterations in 1-carbon metabolism. Overall, our data suggest that maternal prebiotic intake protects offspring against hepatic steatosis and insulin resistance following 21 wk of high fat/sucrose diet, which is in part due to alterations in gut microbiota.-Paul, H. A., Collins, K. H., Nicolucci, A. C., Urbanski, S. J., Hart, D. A., Vogel, H. J., Reimer, R. A. Maternal prebiotic supplementation reduces fatty liver development in offspring through altered microbial and metabolomic profiles in rats.
Collapse
Affiliation(s)
- Heather A Paul
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kelsey H Collins
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | | | - Stefan J Urbanski
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David A Hart
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada; and
| | - Hans J Vogel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Biological Sciences, Bio-Nuclear Magnetic Resonance (NMR) Center, University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
44
|
Baranowski JR, Claud EC. Necrotizing Enterocolitis and the Preterm Infant Microbiome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1125:25-36. [PMID: 30680646 DOI: 10.1007/5584_2018_313] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial colonization patterns in preterm infants differ from those of their term counterparts due to maternal microbial diversity, delivery mode, feeding methods, antibiotic use, and exposure to commensal microbiota and pathogens in the neonatal intensive care unit (NICU). Early gut microbiome dysbiosis predisposes neonates to necrotizing enterocolitis (NEC), a devastating intestinal disease with high morbidity and mortality. Though mechanisms of NEC pathogenesis are not fully understood, the microbiome is a promising therapy target for prevention and treatment. Direct administration of probiotics to preterm infants has been shown to reduce the incidence of NEC, but is not without risk. The immature immune systems of preterm infants leave them vulnerable to even beneficial bacteria. Further research is required to investigate both short-term and long-term effects of probiotic administration to preterm infants. Other methods of altering the preterm infant microbiome must also be considered, including breastfeeding, prebiotics, and targeting the maternal microbiome.
Collapse
Affiliation(s)
| | - Erika C Claud
- Department of Pediatrics, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA.
| |
Collapse
|
45
|
Neuman H, Forsythe P, Uzan A, Avni O, Koren O. Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev 2018; 42:489-499. [PMID: 29945240 DOI: 10.1093/femsre/fuy018] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/23/2018] [Indexed: 12/21/2022] Open
Abstract
Antibiotics are the most common type of medication prescribed to children, including infants, in the Western world. While use of antibiotics has transformed previously lethal infections into relatively minor diseases, antibiotic treatments can have adverse effects as well. It has been shown in children, adults and animal models that antibiotics dramatically alter the gut microbial composition. Since the gut microbiota plays crucial roles in immunity, metabolism and endocrinology, the effects of antibiotics on the microbiota may lead to further health complications. In this review, we present an overview of the effects of antibiotics on the microbiome in children, and correlate them to long-lasting complications of obesity, behavior, allergies, autoimmunity and other diseases.
Collapse
Affiliation(s)
- Hadar Neuman
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel.,Ziv Medical Center, Derech HaRambam St., Safed 13100 Israel.,Zefat Academic College, Jerusalem St. 11, Safed 13206, Israel
| | - Paul Forsythe
- McMaster Brain-Body Institute, St. Joseph's Healthcare Hamilton, L8N 4A6 Hamilton, Ontario, Canada.,Firestone Institute for Respiratory Health and Department of Medicine, 50 Charlton Avenue East, McMaster University, L8N 4A6 Hamilton, Ontario, Canada
| | - Atara Uzan
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Orly Avni
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Omry Koren
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| |
Collapse
|
46
|
Abstract
The developmental origin of health and disease highlights the importance of the period of the first 1000 days (from the conception to the 2 years of life). The process of the gut microbiota establishment is included in this time window. Various perinatal determinants, such as cesarean section delivery, type of feeding, antibiotics treatment, gestational age or environment, can affect the pattern of bacterial colonization and result in dysbiosis. The alteration of the early bacterial gut pattern can persist over several months and may have long-lasting functional effects with an impact on disease risk later in life. As for example, early gut dysbiosis has been involved in allergic diseases and obesity occurrence. Besides, while it was thought that the fetus developed under sterile conditions, recent data suggested the presence of a microbiota in utero, particularly in the placenta. Even if the origin of this microbiota and its eventual transfer to the infant are nowadays unknown, this placental microbiota could trigger immune responses in the fetus and would program the infant's immune development during fetal life, earlier than previously considered. Moreover, several studies demonstrated a link between the composition of placental microbiota and some pathological conditions of the pregnancy. All these data show the evidence of relationships between the neonatal gut establishment and future health outcomes. Hence, the use of pre- and/or probiotics to prevent or repair any early dysbiosis is increasingly attractive to avoid long-term health consequences.
Collapse
|
47
|
Prentice S. They Are What You Eat: Can Nutritional Factors during Gestation and Early Infancy Modulate the Neonatal Immune Response? Front Immunol 2017; 8:1641. [PMID: 29234319 PMCID: PMC5712338 DOI: 10.3389/fimmu.2017.01641] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/09/2017] [Indexed: 12/17/2022] Open
Abstract
The ontogeny of the human immune system is sensitive to nutrition even in the very early embryo, with both deficiency and excess of macro- and micronutrients being potentially detrimental. Neonates are particularly vulnerable to infectious disease due to the immaturity of the immune system and modulation of nutritional immunity may play a role in this sensitivity. This review examines whether nutrition around the time of conception, throughout pregnancy, and in early neonatal life may impact on the developing infant immune system.
Collapse
Affiliation(s)
- Sarah Prentice
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
48
|
Do bacteria shape our development? Crosstalk between intestinal microbiota and HPA axis. Neurosci Biobehav Rev 2017; 83:458-471. [PMID: 28918360 DOI: 10.1016/j.neubiorev.2017.09.016] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/01/2017] [Accepted: 09/12/2017] [Indexed: 02/08/2023]
Abstract
The human body contains as many bacteria in the intestine as the total number of human body cells. These bacteria have a central position in human health and disease, and would also play a role in the regulation of emotions, behavior, and even higher cognitive functions. The Hypothalamic-Pituitary-Adrenal axis (HPA axis) is a major physiological stress system that produces cortisol. This hormone is involved in responding to environmental stress and also shapes many aspects of brain development. Both the HPA axis and the intestinal microbiota show rapid and profound developmental changes during the first years of life. Early environmental disturbances can affect the development of both systems. Early adversity, for example, is known to lead to later unbalances in both, as well as to psychopathological behavior and emotions. The goal of this theoretical review is to summarize current knowledge on the developmental crosstalk between the intestinal microbiota and the HPA axis, providing a basis for understanding the development and bidirectional communication between these two essential systems in human functioning.
Collapse
|