1
|
Khymenets O, Vilarroya O, Benet G, Feixas G, Arranz Betegon A, McLeod MD, Pozo OJ. Profile of steroid metabolites in human breast milk in different stages of lactation. Food Funct 2025. [PMID: 40277187 PMCID: PMC12023736 DOI: 10.1039/d4fo05713a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
Breast milk (BM), as an optimal food, provides the newborn with a variety of minor compounds relevant for health and wellbeing. Endogenous steroids, also minor constituents, are mainly secreted in BM as conjugated metabolites. Recent research has revealed the relevance of steroid conjugates in many physiological processes. Thus, their presence in BM appears to be very intriguing, especially in relation to breastfeeding. The objective of our study was to profile conjugated steroid metabolites present in BM in relation to the lactation stage, and to promote further evaluation of their importance in breastfeeding. For this purpose, we developed and used a direct UHPLC-MS/MS metabolomics approach capable to detect more than 60 conjugated metabolites (mono-sulfated, mono-glucuronylated, bis-sulfated and sulfate-glucuronylated) from all steroid families. We compared the occurrence of these metabolites in samples collected from breastfeeding mothers and stratified by lactation stages: colostrum, transitional and mature milk. Our results showed that many biologically relevant conjugated steroids are secreted in BM. Their concentrations were highest in colostrum, decreased remarkably in transitional and were much lower in mature milk, with some exceptions. The profile of metabolites also differed considerably between lactation stages. The approximate daily secretion in BM indicated that infants are exposed to significant oral doses of steroid conjugates during the first week of lactation. The supply of these metabolites in BM declined and became constant after the second week postpartum. Overall, our data provide a foundation for further investigation on the physiological relevance of BM secreted steroid metabolites in relation to both mother and child.
Collapse
Affiliation(s)
- Olha Khymenets
- Applied Metabolomics Research Group, Neurosciences Research Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| | - Oscar Vilarroya
- Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, Barcelona, Spain
- Neuroimaging Research Group, Neurosciences Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Georgina Benet
- Neuroimaging Research Group, Neurosciences Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Georgina Feixas
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Angela Arranz Betegon
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Malcolm D McLeod
- Research School of Chemistry, Australian National University, Canberra, Australia
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Neurosciences Research Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
2
|
Ryan N, O’Mahony S, Leahy-Warren P, Philpott L, Mulcahy H. The impact of perinatal maternal stress on the maternal and infant gut and human milk microbiomes: A scoping review. PLoS One 2025; 20:e0318237. [PMID: 40019912 PMCID: PMC11870360 DOI: 10.1371/journal.pone.0318237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Perinatal maternal stress, which includes both psychological and physiological stress experienced by healthy women during pregnancy and the postpartum period, is becoming increasingly prevalent. Infant early exposure to adverse environments such as perinatal stress has been shown to increase the long-term risk to metabolic, immunologic and neurobehavioral disorders. Evidence suggests that the human microbiome facilitates the transmission of maternal factors to infants via the vaginal, gut, and human milk microbiomes. The colonization of aberrant microorganisms in the mother's microbiome, influenced by the microbiome-brain-gut axis, may be transferred to infants during a critical early developmental period. This transfer may predispose infants to a more inflammatory-prone microbiome which is associated with dysregulated metabolic process leading to adverse health outcomes. Given the prevalence and potential impact of perinatal stress on maternal and infant health, with no systematic mapping or review of the data to date, the aim of this scoping review is to gather evidence on the relationship between perinatal maternal stress, and the human milk, maternal, and infant gut microbiomes. METHODS This is an exploratory mapping scoping review, guided by the Joanna Briggs Institute's methodology along with use of the Prisma Scr reporting guideline. A comprehensive search was conducted using the following databases, CINAHL Complete; MEDLINE; PsycINFO, Web of Science and Scopus with a protocol registered with Open Science Framework DOI 10.17605/OSF.IO/5SRMV. RESULTS After screening 1145 papers there were 7 paper that met the inclusion criteria. Statistically significant associations were found in five of the studies which identify higher abundance of potentially pathogenic bacteria such as Erwinia, Serratia, T mayombie, Bacteroides with higher maternal stress, and lower levels of stress linked to potentially beneficial bacteria such Lactococcus, Lactobacillus, Akkermansia. However, one study presents conflicting results where it was reported that higher maternal stress was linked to the prevalence of more beneficial bacteria. CONCLUSION This review suggests that maternal stress does have an impact on the alteration of abundance and diversity of influential bacteria in the gut microbiome, however, it can affect colonisation in different ways. These bacterial changes have the capacity to influence long term health and disease. The review analyses data collection tools and methods, offers potential reasons for these findings as well as suggestions for future research.
Collapse
Affiliation(s)
- Niamh Ryan
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| | - Siobhain O’Mahony
- Department of Anatomy and Neuroscience, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Lloyd Philpott
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| | - Helen Mulcahy
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Horwell E, Bearn P, Cutting SM. A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health. MICROBIOME RESEARCH REPORTS 2024; 4:1. [PMID: 40207275 PMCID: PMC11977369 DOI: 10.20517/mrr.2024.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Since the advent of new sequencing and bioinformatic technologies, our understanding of the human microbiome has expanded rapidly over recent years. Numerous studies have indicated causal links between alterations to the microbiome and a range of pathological conditions. Furthermore, a large body of epidemiological data is starting to suggest that exposure, or lack thereof, to specific microbial species during the first five years of life has key implications for long-term health outcomes. These include chronic inflammatory and metabolic conditions such as diabetes, asthma, inflammatory bowel disease (IBD), and obesity, with the effects lasting into adulthood. Human microbial colonisation during these first five years of life is a highly dynamic process, with multiple environmental exposures recently being characterised to have influence before the microbiome stabilises and resembles that of an adult at 3-5 years. This short period of time, known as the window of opportunity, appears to "prime" immunoregulation for later life. Understanding and appreciating this aspect of human physiology is therefore crucial for clinicians, scientists, and public health officials. This review outlines the most recent evidence for the pre- and post-natal environments that order the development of the microbiome, how these influences metabolic and immunoregulatory pathways, and their associated health outcomes. It also discusses the limitations of the current knowledge base, and describes the potential microbiome-mediated interventions and public health measures that may have therapeutic potential in the future.
Collapse
Affiliation(s)
- Edward Horwell
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Philip Bearn
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Simon M. Cutting
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
| |
Collapse
|
4
|
Henriksson HB, Hellström A, Nilsson AK, Sjöbom U, Jönsson B, Frändberg S. Bacterial species in cord blood and their significance in the context of clinical use. Transfus Apher Sci 2024; 63:103961. [PMID: 38981148 DOI: 10.1016/j.transci.2024.103961] [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: 01/10/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/11/2024]
Abstract
Approximately 90 % of infants born before 28 full weeks(extremely-preterm-infants) receive erythrocyte transfusions in early life. Umbilical cord blood(UCB) has been investigated as an alternative source for erythrocyte transfusions to preterm neonates. This retrospective study aimed to compile/evaluate spectrum of bacteria groups/species intermittently detected in processed UCB at National-Swedish-Cord blood bank, (NS-CBB) during the years 2008-2020. Consecutive data from the years 2008-2020 were investigated. UCB from healthy newborns born after 37 full weeks of gestation was collected following clamping of cord (1 min) through cannulation of umbilical vein(vaginal-and C-section-deliveries). In total, 5194 cord blood units (UCBUs) that met NS-CBB-guidelines for total nucleated-cell-content(TNC) were manufactured from 8875 collections. Of 5194 UCBUs,77,6 % were from vaginal-and 22,4 % from C-section deliveries.Samples(10 mL) were collected from surplus eryhtrocyte fraction post-processing(n = 5194), transferred into BACT/ALERT® aerobic/anaerobic culture flasks and monitored 10 days using BACT/ALERT®-3D-Microbial-Detection-Systems. Positive samples were subcultured and typed for bacterial groups and/or species. Out of 5194 processed sampled UCB units,186 (3,6 %) were discarded due to positive sterility tests, 92 % were detected in samples from vaginal-deliveries and 8 % from C-section-deliveries. In all,16 different groups of bacteria and 27 species were identified. Common bacterial/groups and species were anaerobe gram-negative rods(n = 28),coagulase-negative-staphylococci(n = 21),gram-positive rods(n = 21),anaerobe-gram-positive cocci(n = 20) and viridans-streptococci(n = 13). Extracted from these results,in positive samples(n = 13) from C-section deliveries, bacteria were found:viridans-streptococci(n = 7),Aerococcus-urinae(n = 1), Staphylococcus lugdunensis(n = 1),other coagulase-negative staphylococci(n = 1) or a mix of aerobic/anaerobic bacteria(n = 3). Our results are in alignment with previously published contamination rates in processed UCBUs. Still, results point towards importance of strict microbial monitoring when manufacturing UCBUs to achieve patient-safe- products for stem-cell transplantation/transfusion.
Collapse
Affiliation(s)
- Helena Barreto Henriksson
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Research, Development, Education and Innovation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders K Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Sjöbom
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Learning and Leadership for Health Care Professionals at the Institute of Health and Care Science at Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Bodil Jönsson
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sofia Frändberg
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
5
|
Sokou R, Moschari E, Palioura AE, Palioura AP, Mpakosi A, Adamakidou T, Vlachou E, Theodoraki M, Iacovidou N, Tsartsalis AN. The Impact of Gestational Diabetes Mellitus (GDM) on the Development and Composition of the Neonatal Gut Microbiota: A Systematic Review. Microorganisms 2024; 12:1564. [PMID: 39203408 PMCID: PMC11356352 DOI: 10.3390/microorganisms12081564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is an important health issue, as it is connected with adverse effects to the mother as well as the fetus. A factor of essence for the pathology of this disorder is the gut microbiota, which seems to have an impact on the development and course of GDM. The role of the gut microbiota on maternal reproductive health and all the changes that happen during pregnancy as well as during the neonatal period is of high interest. The correct establishment and maturation of the gut microbiota is of high importance for the development of basic biological systems. The aim of this study is to provide a systematic review of the literature on the effect of GDM on the gut microbiota of neonates, as well as possible links to morbidity and mortality of neonates born to mothers with GDM. Systematic research took place in databases including PubMed and Scopus until June 2024. Data that involved demographics, methodology, and changes to the microbiota were derived and divided based on patients with exposure to or with GDM. The research conducted on online databases revealed 316 studies, of which only 16 met all the criteria and were included in this review. Research from the studies showed great heterogeneity and varying findings at the level of changes in α and β diversity and enrichment or depletion in phylum, gene, species, and operational taxonomic units in the neonatal gut microbiota of infants born to mothers with GDM. The ways in which the microbiota of neonates and infants are altered due to GDM remain largely unclear and require further investigation. Future studies are needed to explore and clarify these mechanisms.
Collapse
Affiliation(s)
- Rozeta Sokou
- Neonatal Intensive Care Unit, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece; (E.M.); (A.E.P.); (A.-P.P.); (M.T.)
- Neonatal Department, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Eirini Moschari
- Neonatal Intensive Care Unit, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece; (E.M.); (A.E.P.); (A.-P.P.); (M.T.)
| | - Alexia Eleftheria Palioura
- Neonatal Intensive Care Unit, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece; (E.M.); (A.E.P.); (A.-P.P.); (M.T.)
| | - Aikaterini-Pothiti Palioura
- Neonatal Intensive Care Unit, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece; (E.M.); (A.E.P.); (A.-P.P.); (M.T.)
| | - Alexandra Mpakosi
- Department of Microbiology, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece;
| | - Theodoula Adamakidou
- Department of Nursing, School of Health Sciences, University of West Attica, Ag. Spydironos 28, 12243 Athens, Greece; (T.A.); (E.V.)
| | - Eugenia Vlachou
- Department of Nursing, School of Health Sciences, University of West Attica, Ag. Spydironos 28, 12243 Athens, Greece; (T.A.); (E.V.)
| | - Martha Theodoraki
- Neonatal Intensive Care Unit, General Hospital of Nikea “Agios Panteleimon”, 18454 Piraeus, Greece; (E.M.); (A.E.P.); (A.-P.P.); (M.T.)
| | - Nicoletta Iacovidou
- Neonatal Department, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Athanasios N. Tsartsalis
- Department of Endocrinology Diabetes and Metabolism, Naval Hospital of Athens, Dinokratous 70, 11521 Athens, Greece;
| |
Collapse
|
6
|
Jara J, Alba C, Del Campo R, Fernández L, Sáenz de Pipaón M, Rodríguez JM, Orgaz B. Linking preterm infant gut microbiota to nasograstric enteral feeding tubes: exploring potential interactions and microbial strain transmission. Front Pediatr 2024; 12:1397398. [PMID: 38952433 PMCID: PMC11215057 DOI: 10.3389/fped.2024.1397398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction Preterm birth is a growing problem worldwide. Staying at a neonatal intensive care unit (NICU) after birth is critical for the survival of preterm infants whose feeding often requires the use of nasogastric enteral feeding tubes (NEFT). These can be colonized by hospital-associated pathobionts that can access the gut of the preterm infants through this route. Since the gut microbiota is the most impactful factor on maturation of the immune system, any disturbance in this may condition their health. Therefore, the aim of this study is to assess the impact of NEFT-associated microbial communities on the establishment of the gut microbiota in preterm infants. Material and methods A metataxonomic analysis of fecal and NEFT-related samples obtained during the first 2 weeks of life of preterm infants was performed. The potential sharing of strains isolated from the same set of samples of bacterial species involved in NICU's outbreaks, was assessed by Random Amplification of Polymorphic DNA (RAPD) genotyping. Results In the samples taken 48 h after birth (NEFT-1 and Me/F1), Staphylococcus spp. was the most abundant genera (62% and 14%, respectively) and it was latter displaced to 5.5% and 0.45%, respectively by Enterobacteriaceae. Significant differences in beta diversity were detected in NEFT and fecal samples taken at day 17 after birth (NEFT-3 and F3) (p = 0.003 and p = 0.024, respectively). Significant positive correlations were found between the most relevant genera detected in NEFT-3 and F3. 28% of the patients shared at least one RAPD-PCR profile in fecal and NEFT samples and 11% of the total profiles were found at least once simultaneously in NEFT and fecal samples from the same patient. Conclusion The results indicate a parallel bacterial colonization of the gut of preterm neonates and the NEFTs used for feeding, potentially involving strain sharing between these niches. Moreover, the same bacterial RAPD profiles were found in neonates hospitalized in different boxes, suggesting a microbial transference within the NICU environment. This study may assist clinical staff in implementing best practices to mitigate the spread of pathogens that could threaten the health of preterm infants.
Collapse
Affiliation(s)
- J. Jara
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - C. Alba
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - R. Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - L. Fernández
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - M. Sáenz de Pipaón
- Department of Neonatology, La Paz University Hospital of Madrid, Madrid, Spain
- Department of Pediatrics, Autonoma University of Madrid, Madrid, Spain
| | - J. M. Rodríguez
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - B. Orgaz
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| |
Collapse
|
7
|
Han YZ, Du BX, Zhu XY, Wang YZY, Zheng HJ, Liu WJ. Lipid metabolism disorder in diabetic kidney disease. Front Endocrinol (Lausanne) 2024; 15:1336402. [PMID: 38742197 PMCID: PMC11089115 DOI: 10.3389/fendo.2024.1336402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic kidney disease (DKD), a significant complication associated with diabetes mellitus, presents limited treatment options. The progression of DKD is marked by substantial lipid disturbances, including alterations in triglycerides, cholesterol, sphingolipids, phospholipids, lipid droplets, and bile acids (BAs). Altered lipid metabolism serves as a crucial pathogenic mechanism in DKD, potentially intertwined with cellular ferroptosis, lipophagy, lipid metabolism reprogramming, and immune modulation of gut microbiota (thus impacting the liver-kidney axis). The elucidation of these mechanisms opens new potential therapeutic pathways for DKD management. This research explores the link between lipid metabolism disruptions and DKD onset.
Collapse
Affiliation(s)
- Yi-Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing-Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang-Zhi-Yuan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei-Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Govindarajan V, Devadas S, Shah PA, Diggikar S. Impact of Kangaroo Mother Care on Skin Microbiome of Very Preterm Infants - A Pilot Study. Indian J Pediatr 2024; 91:229-234. [PMID: 37040016 DOI: 10.1007/s12098-023-04562-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/10/2023] [Indexed: 04/12/2023]
Abstract
OBJECTIVES To test whether Kangaroo mother care (KMC) aids in transfer of favourable skin microbiome from mother to infant by comparing the microbiome composition before and after KMC. METHODS A prospective cohort pilot study was conducted in a Level III neonatal intensive care unit (NICU) in South India, recruiting 30 preterm infants with gestation <32 wk from October 2020 through December 2020. Neonatal skin involving the area in contact with the mother during KMC i.e., axilla, chest and abdomen was swabbed at the end of first week of life, prior to initiation of KMC. The 2nd swab involving the same areas was taken following KMC for 7 d for at least 6 h a day. The swabs were analysed using Next Generation Sequencing (NGS) - 16sRNA and abundance of organisms isolated were mapped. Statistical analyses using t-test and PERMANOVA were performed to compare phyla and genera of bacterial abundance pre-KMC and post-KMC. RESULTS KMC at phyla level increased the relative abundance of Firmicutes (p=0.52) and significantly decreased Proteobacteria (p=0.02). At species level, KMC decreased pathogenic bacterial count of Escherichia (p=0.05), while counts of S. hemolyticus (p=0.01) and S. hominis (p=.002) significantly increased post KMC. CONCLUSIONS KMC has a potential role in altering the neonatal skin microbiota towards a more favourable microenvironment. The clinical significance of these novel findings needs to be validated with larger studies.
Collapse
Affiliation(s)
- Varun Govindarajan
- Department of Pediatrics, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Sahana Devadas
- Department of Pediatrics, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Pritik A Shah
- Undergraduate (MBBS), Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Shivashankar Diggikar
- Department of Pediatrics, Oyster Woman and Child Hospital, Bengaluru, Karnataka, India.
| |
Collapse
|
9
|
Kumar B, Lorusso E, Fosso B, Pesole G. A comprehensive overview of microbiome data in the light of machine learning applications: categorization, accessibility, and future directions. Front Microbiol 2024; 15:1343572. [PMID: 38419630 PMCID: PMC10900530 DOI: 10.3389/fmicb.2024.1343572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Metagenomics, Metabolomics, and Metaproteomics have significantly advanced our knowledge of microbial communities by providing culture-independent insights into their composition and functional potential. However, a critical challenge in this field is the lack of standard and comprehensive metadata associated with raw data, hindering the ability to perform robust data stratifications and consider confounding factors. In this comprehensive review, we categorize publicly available microbiome data into five types: shotgun sequencing, amplicon sequencing, metatranscriptomic, metabolomic, and metaproteomic data. We explore the importance of metadata for data reuse and address the challenges in collecting standardized metadata. We also, assess the limitations in metadata collection of existing public repositories collecting metagenomic data. This review emphasizes the vital role of metadata in interpreting and comparing datasets and highlights the need for standardized metadata protocols to fully leverage metagenomic data's potential. Furthermore, we explore future directions of implementation of Machine Learning (ML) in metadata retrieval, offering promising avenues for a deeper understanding of microbial communities and their ecological roles. Leveraging these tools will enhance our insights into microbial functional capabilities and ecological dynamics in diverse ecosystems. Finally, we emphasize the crucial metadata role in ML models development.
Collapse
Affiliation(s)
- Bablu Kumar
- Università degli Studi di Milano, Milan, Italy
- Department of Biosciences, Biotechnology and Environment, University of Bari A. Moro, Bari, Italy
| | - Erika Lorusso
- Department of Biosciences, Biotechnology and Environment, University of Bari A. Moro, Bari, Italy
- National Research Council, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| | - Bruno Fosso
- Department of Biosciences, Biotechnology and Environment, University of Bari A. Moro, Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology and Environment, University of Bari A. Moro, Bari, Italy
- National Research Council, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| |
Collapse
|
10
|
Gao C, Koko MY, Hong W, Gankhuyag J, Hui M, Gantumur MA, Dong N. Protective Properties of Intestinal Alkaline Phosphatase Supplementation on the Intestinal Barrier: Interactions and Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27-45. [PMID: 37964463 DOI: 10.1021/acs.jafc.3c05119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
The intestinal barrier is critical for maintaining intestinal homeostasis, and its dysfunction is associated with various diseases. Recent findings have revealed the multifunctional role of intestinal alkaline phosphatase (IAP) in diverse biological processes, including gut health maintenance and function. This review summarizes the protective effects of IAP on intestinal barrier integrity, encompassing the physical, chemical, microbial, and immune barriers. We discuss the results and insights from in vitro, animal model, and clinical studies as well as the available evidence regarding the impact of diet on IAP activity and expression. IAP can also be used as an indicator to assess intestinal-barrier-related diseases. Further research into the mechanisms of action and long-term health effects of IAP in maintaining overall intestinal health is essential for its future use as a dietary supplement or functional component in medical foods.
Collapse
Affiliation(s)
- Chenzhe Gao
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Marwa Yagoub Koko
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Weichen Hong
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Javzan Gankhuyag
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Mizhou Hui
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Munkh-Amgalan Gantumur
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Na Dong
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
| |
Collapse
|
11
|
Han YZ, Zheng HJ, Du BX, Zhang Y, Zhu XY, Li J, Wang YX, Liu WJ. Role of Gut Microbiota, Immune Imbalance, and Allostatic Load in the Occurrence and Development of Diabetic Kidney Disease. J Diabetes Res 2023; 2023:8871677. [PMID: 38094870 PMCID: PMC10719010 DOI: 10.1155/2023/8871677] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic kidney disease (DKD) is a prevailing complication arising from diabetes mellitus. Unfortunately, there are no trustworthy and efficacious treatment modalities currently available. In recent times, compelling evidence has emerged regarding the intricate correlation between the kidney and the gut microbiota, which is considered the largest immune organ within the human physique. Various investigations have demonstrated that the perturbation of the gut microbiota and its associated metabolites potentially underlie the etiology and progression of DKD. This phenomenon may transpire through perturbation of both the innate and the adaptive immunity, leading to a burdensome allostatic load on the body and ultimately culminating in the development of DKD. Within this literature review, we aim to delve into the intricate interplay between the gut microbiota, its metabolites, and the immune system in the context of DKD. Furthermore, we strive to explore and elucidate potential chemical interventions that could hold promise for the treatment of DKD, thereby offering invaluable insights and directions for future research endeavors.
Collapse
Affiliation(s)
- Yi Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Li
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yao Xian Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Cheddadi R, Yeramilli V, Martin C. From Mother to Infant, from Placenta to Gut: Understanding Varied Microbiome Profiles in Neonates. Metabolites 2023; 13:1184. [PMID: 38132866 PMCID: PMC10745069 DOI: 10.3390/metabo13121184] [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: 11/03/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
The field of human microbiome and gut microbial diversity research has witnessed a profound transformation, driven by advances in omics technologies. These advancements have unveiled essential connections between microbiome alterations and severe conditions, prompting the development of new frameworks through epidemiological studies. Traditionally, it was believed that each individual harbored unique microbial communities acquired early in life, evolving over the course of their lifetime, with little acknowledgment of any prenatal microbial development, but recent research challenges this belief. The neonatal microbiome's onset, influenced by factors like delivery mode and maternal health, remains a subject of intense debate, hinting at potential intrauterine microbial processes. In-depth research reveals associations between microbiome profiles and specific health outcomes, ranging from obesity to neurodevelopmental disorders. Understanding these diverse microbiome profiles is essential for unraveling the intricate relationships between the microbiome and health outcomes.
Collapse
Affiliation(s)
- Riadh Cheddadi
- Department of Surgery, Division of Pediatric Surgery, Washington University School of Medicine, Saint Louis, MO 63110, USA (C.M.)
| | | | | |
Collapse
|
13
|
Stanikova A, Jouza M, Bohosova J, Slaby O, Jabandziev P. Role of the microbiome in pathophysiology of necrotising enterocolitis in preterm neonates. BMJ Paediatr Open 2023; 7:e002172. [PMID: 37918941 PMCID: PMC10626796 DOI: 10.1136/bmjpo-2023-002172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Although necrotising enterocolitis (NEC) is a serious, life-threatening disease, improved neonatal care is increasing the number of survivors with NEC among extremely preterm neonates. Therapy is nevertheless mostly symptomatic and the mortality rate remains high, especially among neonates requiring surgery. Therefore, it is important to focus on preventing the disease and modifiable risk factors. NEC's pathophysiology is multifaceted, with key factors being immaturity of the immune and barrier protective mechanisms of the premature gut and exaggerated proinflammatory reaction to insults like gut hypoxia, enteral nutrition or microbial dysbiosis. The role of the intestinal microbiome in the pathophysiology of NEC has been a subject of research for many years, but to date no specific pathogen or type of dysbiosis has been connected with NEC development. This review assesses current knowledge as to the role of the intestinal microbiota in the pathophysiology of NEC and the possibilities for positively influencing it.
Collapse
Affiliation(s)
- Andrea Stanikova
- Department of Neonatology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Jouza
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| | - Julia Bohosova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, University Hospital Brno, Brno, Czech Republic
| | - Petr Jabandziev
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
14
|
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
|
15
|
Panzer JJ, Romero R, Greenberg JM, Winters AD, Galaz J, Gomez-Lopez N, Theis KR. Is there a placental microbiota? A critical review and re-analysis of published placental microbiota datasets. BMC Microbiol 2023; 23:76. [PMID: 36934229 PMCID: PMC10024458 DOI: 10.1186/s12866-023-02764-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/10/2023] [Indexed: 03/20/2023] Open
Abstract
The existence of a placental microbiota is debated. The human placenta has historically been considered sterile and microbial colonization was associated with adverse pregnancy outcomes. Yet, recent DNA sequencing investigations reported a microbiota in typical human term placentas. However, this detected microbiota could represent background DNA or delivery-associated contamination. Using fifteen publicly available 16S rRNA gene datasets, existing data were uniformly re-analyzed with DADA2 to maximize comparability. While Amplicon Sequence Variants (ASVs) identified as Lactobacillus, a typical vaginal bacterium, were highly abundant and prevalent across studies, this prevalence disappeared after applying likely DNA contaminant removal to placentas from term cesarean deliveries. A six-study sub-analysis targeting the 16S rRNA gene V4 hypervariable region demonstrated that bacterial profiles of placental samples and technical controls share principal bacterial ASVs and that placental samples clustered primarily by study origin and mode of delivery. Contemporary DNA-based evidence does not support the existence of a placental microbiota.ImportanceEarly-gestational microbial influences on human development are unclear. By applying DNA sequencing technologies to placental tissue, bacterial DNA signals were observed, leading some to conclude that a live bacterial placental microbiome exists in typical term pregnancy. However, the low-biomass nature of the proposed microbiome and high sensitivity of current DNA sequencing technologies indicate that the signal may alternatively derive from environmental or delivery-associated bacterial DNA contamination. Here we address these alternatives with a re-analysis of 16S rRNA gene sequencing data from 15 publicly available placental datasets. After identical DADA2 pipeline processing of the raw data, subanalyses were performed to control for mode of delivery and environmental DNA contamination. Both environment and mode of delivery profoundly influenced the bacterial DNA signal from term-delivered placentas. Aside from these contamination-associated signals, consistency was lacking across studies. Thus, placentas delivered at term are unlikely to be the original source of observed bacterial DNA signals.
Collapse
Affiliation(s)
- Jonathan J Panzer
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA.
- Detroit Medical Center, Detroit, Michigan, USA.
| | - Jonathan M Greenberg
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Andrew D Winters
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Kevin R Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, USA.
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.
| |
Collapse
|
16
|
Odakura AM, Caldara FR, Burbarelli MFDC, Almeida Paz ICDL, Garcia RG, Oliveira dos Santos VM, de Brito Mandu DF, Braz JM, Lourenço da Silva MI. Dietary Supplementation of Eubiotic Fiber Based on Lignocellulose on Performance and Welfare of Gestating and Lactating Sows. Animals (Basel) 2023; 13:ani13040695. [PMID: 36830483 PMCID: PMC9952275 DOI: 10.3390/ani13040695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
The present study aimed to evaluate the effects of partially fermentable insoluble dietary fiber supplementation on the behavior, surface temperature, and reproductive parameters of gestating and lactating sows, as well as on the performance of their litters. Four hundred hyper-prolific sows were assigned in a randomized block design with two treatment groups during the gestation phase: Control (C), corn-soy based diets, or corn-soy based diets with daily 55 g supplementation of eubiotic fiber (F) from the 85th day of gestation until the farrowing (late pregnancy). During the lactation phase, the sows were assigned in a 2 × 2 factorial design using the following treatment groups: (1) CC, no fiber included during gestation and lactation. (2) FC, daily inclusion of 55 g of fiber only during late pregnancy. (3) CF, daily inclusion of 55 g of fiber only during lactation. (4) FF, daily inclusion of 55 g of fiber during late pregnancy and lactation. Sows that received dietary fiber supplementation during the final third of gestation increased feed intake during lactation. Piglets from sows supplemented in both phases showed a significant increase in weight at weaning. The dietary supplementation of eubiotic fiber for sows in the end period of gestation and lactation improved performance and welfare, with positive consequences for developing their litters.
Collapse
Affiliation(s)
- Agnês Markiy Odakura
- College of Agrarian Sciences (FCA), Federal University of Grande Dourados (UFGD), Dourados 79804-970, Brazil
- Correspondence:
| | - Fabiana Ribeiro Caldara
- College of Agrarian Sciences (FCA), Federal University of Grande Dourados (UFGD), Dourados 79804-970, Brazil
| | | | | | - Rodrigo Garófallo Garcia
- College of Agrarian Sciences (FCA), Federal University of Grande Dourados (UFGD), Dourados 79804-970, Brazil
| | - Viviane Maria Oliveira dos Santos
- Faculty of Veterinary Medicine and Animal Sciences (FAMEZ), Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | | | - Jaqueline Murback Braz
- College of Agrarian Sciences (FCA), Federal University of Grande Dourados (UFGD), Dourados 79804-970, Brazil
| | | |
Collapse
|
17
|
Vemuri R, Herath MP. Beyond the Gut, Emerging Microbiome Areas of Research: A Focus on Early-Life Microbial Colonization. Microorganisms 2023; 11:microorganisms11020239. [PMID: 36838204 PMCID: PMC9962807 DOI: 10.3390/microorganisms11020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Undoubtedly, the human body harbors trillions of microbes of different kinds performing various physiological activities, such as priming the immune system, influencing host metabolism, and improving health by providing important metabolites such as short-chain fatty acids. Although the gut is considered the "microbial organ" of our body as it hosts the most microbes, there are microbes present in various other important anatomical locations differing in numbers and type. Research has shown the presence of microbes in utero, sparking a debate on the "sterile womb" concept, and there is much scope for more work in this area. It is important to understand the early-life microbiome colonization, which has a role in the developmental origins of health and disease in later life. Moreover, seminal studies have indicated the presence of microbes beyond the gut, for example, in the adipose tissue and the liver. However, it is still unclear what is the exact source of these microbes and their exact roles in health and disease. In this review, we appraise and discuss emerging microbiome areas of research and their roles in metabolic health. Further, we review the importance of the genital microbiome in early-life microbial interactions.
Collapse
Affiliation(s)
- Ravichandra Vemuri
- Department of Pathology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
- Correspondence: (R.V.); (M.P.H.)
| | - Manoja P. Herath
- School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia
- Correspondence: (R.V.); (M.P.H.)
| |
Collapse
|
18
|
Neonatal Healthcare-Associated Conjunctivitis: A Descriptive Study from Saudi Arabia. Medicina (B Aires) 2022; 58:medicina58101448. [PMID: 36295608 PMCID: PMC9608085 DOI: 10.3390/medicina58101448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Healthcare-associated conjunctivitis (HAC) has been associated with serious ophthalmological complications in neonates, including blindness. This three-year retrospective, descriptive study was conducted between 2019 and 2021 to determine the most common bacteria associated with neonatal HAC at a tertiary-care hospital in Saudi Arabia. The inclusion criteria were defined based on the centers for disease control and prevention (CDC) guidelines for the diagnosis of neonatal HAC. When HAC was clinically suspected, conjunctival swabs were obtained from neonates and sent to the microbiology lab following standard protocols. A univariate analysis was conducted on the included samples. A total of 79 cases met our inclusion criteria and were retrospectively studied. A descriptive analysis showed that Pseudomonas aeruginosa was the leading cause of HAC, with 25% (20 cases), followed by Escherichia coli and Klebsiella pneumonia (11.5% for each). About 9% of the analyzed cases were positive for Staphylococcus aureus. Orogastric feeding was the most commonly (94%) associated factor with HAC, followed by respiratory distress syndrome (RDS) and preterm birth, which were found in 70% and 64% of the cases, respectively. To conclude, HAC is an alarming healthcare problem, and bacteria, including Gram-negative bacteria, are common causes. Thus, physician awareness, effective communication with microbiologists, and the implementation of infection control recommendations, including hand hygiene, could minimize this problem and avoid the serious complications of HAC.
Collapse
|
19
|
Pereira MDL, Levy M, Nissapatorn V, de Oliveira GLV. Editorial: Women in microbiome in health and disease 2021. Front Cell Infect Microbiol 2022; 12:1054190. [PMID: 36304933 PMCID: PMC9593082 DOI: 10.3389/fcimb.2022.1054190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Maria de Lourdes Pereira
- Centre for Research in Ceramics and Composite Materials (CICECO) - Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Maayan Levy
- Microbiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and World Union for Herbal Drug Discovery [WUHeDD], Walailak University, Nakhon Si Thammarat, Thailand
- *Correspondence: Gislane Lelis Vilela de Oliveira, ; Veeranoot Nissapatorn,
| | - Gislane Lelis Vilela de Oliveira
- Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), Sao Jose do Rio Preto, Brazil
- *Correspondence: Gislane Lelis Vilela de Oliveira, ; Veeranoot Nissapatorn,
| |
Collapse
|
20
|
Zakošek Pipan M, Podpečan O, Mrkun J. The fascinating microbes and their impact on neonatal dogs and cats - A review. Acta Vet Hung 2022; 70:175-183. [PMID: 35976733 DOI: 10.1556/004.2022.00022] [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: 06/02/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022]
Abstract
Recent literature data indicate that canine and feline neonates are not born in a sterile environment as it was stated previously. The acquisition, colonisation and maintenance of the early life microbiota of healthy fetuses is a rapidly developing research area. In humans, the natural healthy infant microbiome plays an essential role in health and its assembly is determined by the maternal-offspring exchanges of microbes. Even though this topic is becoming more and more important in dogs and cats, the exact role of the neonatal microbiome is not yet fully known in animals. This review summarises the current knowledge of the normal physiological neonatal microbiome in healthy puppies and kittens.
Collapse
Affiliation(s)
- Maja Zakošek Pipan
- Clinic for Reproduction and Large Animals, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Ožbalt Podpečan
- Clinic for Reproduction and Large Animals, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Janko Mrkun
- Clinic for Reproduction and Large Animals, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
21
|
Positive Effects of α-Lactalbumin in the Management of Symptoms of Polycystic Ovary Syndrome. Nutrients 2022; 14:nu14153220. [PMID: 35956395 PMCID: PMC9370664 DOI: 10.3390/nu14153220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
To date, the involvement of α-Lactalbumin (α-LA) in the management of polycystic ovary syndrome (PCOS) refers to its ability to improve intestinal absorption of natural molecules like inositols, overcoming the inositol resistance. However, due to its own aminoacidic building blocks, α-LA is involved in various biological processes that can open new additional applications. A great portion of women with PCOS exhibit gastrointestinal dysbiosis, which is in turn one of the triggering mechanisms of the syndrome. Due to its prebiotic effect, α-LA can recover dysbiosis, also improving the insulin resistance, obesity and intestinal inflammation frequently associated with PCOS. Further observations suggest that altered gut microbiota negatively influence mental wellbeing. Depressive mood and low serotonin levels are indeed common features of women with PCOS. Thanks to its content of tryptophan, which is the precursor of serotonin, and considering the strict link between gut and brain, using α-LA contributes to preserving mental well-being by maintaining high levels of serotonin. In addition, considering women with PCOS seeking pregnancy, both altered microbiota and serotonin levels can induce later consequences in the offspring. Therefore, a deeper knowledge of potential applications of α-LA is required to transition to preclinical and clinical studies extending its therapeutic advantages in PCOS.
Collapse
|
22
|
Nathan AM, Chong KN, Teh CSJ, Hng SY, Eg KP, de Bruyne JA, Muhamad AN, Adam Q, Zaki RA, Razali N. Colonization of the Newborn Respiratory Tract and Its Association with Respiratory Morbidity in the first six months of life: A Prospective Cohort Study. Int J Infect Dis 2022; 122:712-720. [PMID: 35843493 DOI: 10.1016/j.ijid.2022.06.049] [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: 06/03/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE We aimed to determine the association between newborn bacterial colonization and infant respiratory morbidity, in the first six months of life. METHODS This prospective study included healthy newborn infants. Nasopharyngeal swabs performed within 72hrs of delivery were analyzed via polymerase chain reaction. We assessed cumulative respiratory morbidity of infants at 6-months-old. RESULTS Four hundred and twenty-six mother-infant pairs were recruited. In 53.3% (n=225) of newborns, S. pneumoniae (46%) and S. aureus (7.3%) was isolated. None had H. influenzae nor M. catarrhalis. At 6-months-old, 50.7% had experienced respiratory symptoms, 25% had unscheduled doctor visits, and 10% were treated with nebulizers. Colonization with S.pneumoniae was associated with reduced risk of any respiratory symptom (aOR 0.39[95% CI 0.16,0.50]), unscheduled doctor visits (aOR 0.35 [95% CI 0.18,0.67]) and nebulizer treatment (aOR 0.23 [95% CI 0.07,0.72]) at 6 months. Pregnancy-induced hypertension was also associated with increased need for nebulizer treatment (aOR 9.11 [95% CI 1.43,58.1]). CONCLUSION Colonization of the newborn respiratory tract occurred in 53% of infants. Streptococcus pneumoniae was the most common organism, and this was associated with a reduced risk for respiratory morbidity at six months of life.
Collapse
Affiliation(s)
- Anna Marie Nathan
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Kai Ning Chong
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Cindy Shuan Ju Teh
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Shih Ying Hng
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kah Peng Eg
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jessie Anne de Bruyne
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Anis Najwa Muhamad
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Quraisiah Adam
- Department of Paediatrics, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Rafdzah Ahmad Zaki
- Center for Epidemiology and Evidence-Based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nuguelis Razali
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Rosa F, Sharma AK, Gurung M, Casero D, Matazel K, Bode L, Simecka C, Elolimy AA, Tripp P, Randolph C, Hand TW, Williams KD, LeRoith T, Yeruva L. Human Milk Oligosaccharides Impact Cellular and Inflammatory Gene Expression and Immune Response. Front Immunol 2022; 13:907529. [PMID: 35844612 PMCID: PMC9278088 DOI: 10.3389/fimmu.2022.907529] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Human milk harbors complex carbohydrates, including human milk oligosaccharides (HMOs), the third most abundant component after lactose and lipids. HMOs have been shown to impact intestinal microbiota, modulate the intestinal immune response, and prevent pathogenic bacterial binding by serving as decoy receptors. However, the direct effect of HMOs on intestinal function and immunity remains to be elucidated. To address this knowledge gap, 21-day-old germ-free mice (C57BI/6) were orally gavaged with 15 mg/day of pooled HMOs for 7 or 14 days and euthanized at day 28 or 35. A set of mice was maintained until day 50 to determine the persistent effects of HMOs. Control groups were maintained in the isolators for 28, 35, or 50 days of age. At the respective endpoints, intestinal tissues were subjected to histomorphometric and transcriptomic analyses, while the spleen and mesenteric lymph nodes (MLNs) were subjected to flow cytometric analysis. The small intestine (SI) crypt was reduced after HMO treatment relative to control at days 28 and 35, while the SI villus height and large intestine (LI) gland depth were decreased in the HMO-treated mice relative to the control at day 35. We report significant HMO-induced and location-specific gene expression changes in host intestinal tissues. HMO treatment significantly upregulated genes involved in extracellular matrix, protein ubiquitination, nuclear transport, and mononuclear cell differentiation. CD4+ T cells were increased in both MLNs and the spleen, while CD8+ T cells were increased in the spleen at day 50 in the HMO group in comparison to controls. In MLNs, plasma cells were increased in HMO group at days 28 and 35, while in the spleen, only at day 28 relative to controls. Macrophages/monocytes and neutrophils were lower in the spleen of the HMO group at days 28, 35, and 50, while in MLNs, only neutrophils were lower at day 50 in the 14-day HMO group. In addition, diphtheria toxoid and tetanus toxoid antibody-secreting cells were higher in HMO-supplemented group compared to controls. Our data suggest that HMOs have a direct effect on gastrointestinal tract metabolism and the immune system even in the absence of host microbiota.
Collapse
Affiliation(s)
- Fernanda Rosa
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Ashok K. Sharma
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai, Los Angeles, CA, United States
| | - Manoj Gurung
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - David Casero
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai, Los Angeles, CA, United States
| | - Katelin Matazel
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Christy Simecka
- Division of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ahmed A. Elolimy
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
- Animal Production Department, National Research Centre, Giza, Egypt
| | - Patricia Tripp
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| | - Christopher Randolph
- Center for Translational Pediatric Research, Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Timothy W. Hand
- University of Pittsburgh School of Medicine, R.K. Mellon Foundation Institute for Pediatric Research, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Keith D. Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences & Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Laxmi Yeruva
- Arkansas Children’s Nutrition Center, United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Little Rock, AR, United States
| |
Collapse
|
24
|
Metagenomic profiles of the early life microbiome of Indonesian inpatient neonates and their influence on clinical characteristics. Sci Rep 2022; 12:9413. [PMID: 35672441 PMCID: PMC9174262 DOI: 10.1038/s41598-022-13496-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/25/2022] [Indexed: 11/21/2022] Open
Abstract
Determining the initial normal neonatal gut microbiome is challenging. The debate regarding the sterile fetal environment is still ongoing. Therefore, studying and comparing normal and dysbiotic microbiomes requires the elucidation of both the fetal and infant microbiomes. Factors influencing the normal microbiome also include regional and genetic factors specific to different countries. Determining the normal microbiome population in our center and their association with the clinical conditions of infants is helpful as a tool for both the prevention and treatment of related diseases during neonatal care. Here, we employed metagenomic sequencing to characterize meconium and the subsequent early-life gut microbiome of preterm neonates in Jakarta, Indonesia. Microbiome diversity and complexity was higher in the meconium and on day 4 than on day 7. At the genus level, the most abundant genus overall was unidentified Enterobacteriaceae, with meconium samples dominated by Ureaplasma, day 4 fecal samples dominated by Staphylococcus, and day 7 samples dominated by Clostridiales, while at the phylum level the most abundant was Proteobacteria and Firmicutes. Perinatal factors of PROM and mother’s diet influenced the meconium microbiome, while day 4 and day 7 microbiome was associated with bacteremia and early administration of antibiotics. One of our sample sets was derived from triplets, and they had varying diversity despite being triplets. These data are valuable for understanding the formation of a healthy microbiome specific to neonates and devising a strategy to improve both the gut health and related clinical outcomes of the neonate.
Collapse
|
25
|
Breastfeeding as a regulating factor of the development of the intestinal microbiome in the early stages of life. Eur Food Res Technol 2022. [DOI: 10.1007/s00217-022-04012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Ramírez-Lozada T, Loranca-García MC, Fuentes-Venado CE, Rodríguez-Cerdeira C, Ocharan-Hernández E, Soriano-Ursúa MA, Farfán-García ED, Chávez-Gutiérrez E, Ramírez-Magaña X, Robledo-Cayetano M, Loza-Mejía MA, Santa-Olalla IAG, Torres-Paez OU, Pinto-Almazán R, Martínez-Herrera E. Does the Fetus Limit Antibiotic Treatment in Pregnant Patients with COVID-19? Antibiotics (Basel) 2022; 11:252. [PMID: 35203854 PMCID: PMC8868538 DOI: 10.3390/antibiotics11020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
During pregnancy, there is a state of immune tolerance that predisposes them to viral infection, causing maternal-fetal vulnerability to the adverse effects of COVID-19. Bacterial coinfections significantly increase the mortality rate for COVID-19. However, it is known that all drugs, including antibiotics, will enter the fetal circulation in a variable degree despite the role of the placenta as a protective barrier and can cause teratogenesis or other malformations depending on the timing of exposure to the drug. Also, it is important to consider the impact of the indiscriminate use of antibiotics during pregnancy can alter both the maternal and fetal-neonatal microbiota, generating future repercussions in both. In the present study, the literature for treating bacterial coinfections in pregnant women with COVID-19 is reviewed. In turn, we present the findings in 50 pregnant women hospitalized diagnosed with SARS-CoV-2 without previous treatment with antibiotics; moreover, a bacteriological culture of sample types was performed. Seven pregnant women had coinfection with Staphylococcus haemolyticus, Staphylococcus epidermidis, Streptococcus agalactiae, Escherichia coli ESBL +, biotype 1 and 2, Acinetobacter jahnsonii, Enterococcus faecium, and Clostridium difficile. When performing the antibiogram, resistance to multiple drugs was found, such as macrolides, aminoglycosides, sulfa, dihydrofolate reductase inhibitors, beta-lactams, etc. The purpose of this study was to generate more scientific evidence on the better use of antibiotics in these patients. Because of this, it is important to perform an antibiogram to prevent abuse of empirical antibiotic treatment with antibiotics in pregnant women diagnosed with SARS-CoV-2.
Collapse
Affiliation(s)
- Tito Ramírez-Lozada
- Servicio de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca 56530, Mexico; (T.R.-L.); (X.R.-M.)
| | - María Concepción Loranca-García
- Hospital General de Zona No. 53, Los Reyes, Instituto Mexicano del Seguro Social (IMSS), Carr Federal México-Puebla Km 17.5, Villa de la Paz, Rincón de los Reyes, Los Reyes Acaquilpan 56400, Mexico;
| | | | - Carmen Rodríguez-Cerdeira
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain;
- Dermatology Department, Hospital Vithas Ntra. Sra. de Fátima, 36206 Vigo, Spain
- Campus Universitario, University of Vigo, 36310 Vigo, Spain
| | - Esther Ocharan-Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (E.O.-H.); (M.A.S.-U.); (E.D.F.-G.)
| | - Marvin A. Soriano-Ursúa
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (E.O.-H.); (M.A.S.-U.); (E.D.F.-G.)
| | - Eunice D. Farfán-García
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (E.O.-H.); (M.A.S.-U.); (E.D.F.-G.)
| | - Edwin Chávez-Gutiérrez
- Doctorado en Ciencias en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, IPN, Mexico City 07738, Mexico;
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de Mexico 56530, Mexico; (M.R.-C.); (O.U.T.-P.)
| | - Xóchitl Ramírez-Magaña
- Servicio de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca 56530, Mexico; (T.R.-L.); (X.R.-M.)
| | - Maura Robledo-Cayetano
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de Mexico 56530, Mexico; (M.R.-C.); (O.U.T.-P.)
| | - Marco A. Loza-Mejía
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico;
| | | | - Oscar Uriel Torres-Paez
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Estado de Mexico 56530, Mexico; (M.R.-C.); (O.U.T.-P.)
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (E.O.-H.); (M.A.S.-U.); (E.D.F.-G.)
- Non-Communicable Disease Research Group, Facultad Mexicana de Medicina, Universidad La Salle-México, Las Fuentes 17, Tlalpan Centro I, Tlalpan, Mexico City 14000, Mexico
| | - Erick Martínez-Herrera
- Efficiency, Quality, and Costs in Health Services Research Group (EFISALUD), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (E.O.-H.); (M.A.S.-U.); (E.D.F.-G.)
| |
Collapse
|
27
|
Cheng Y, Selma-Royo M, Cao X, Calatayud M, Qi Q, Zhou J, Zeng L, Garcia-Mantrana I, Collado MC, Han B. Influence of Geographical Location on Maternal-Infant Microbiota: Study in Two Populations From Asia and Europe. Front Cell Infect Microbiol 2022; 11:663513. [PMID: 35186776 PMCID: PMC8855098 DOI: 10.3389/fcimb.2021.663513] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 12/21/2021] [Indexed: 01/01/2023] Open
Abstract
Early gut microbial colonization is driven by many factors, including mode of birth, breastfeeding, and other environmental conditions. Characters of maternal-neonatal microbiota were analyzed from two distinct populations in similar latitude but different continents (Oriental Asia and Europe). A total number of 120 healthy families from China (n=60) and Spain (n=60) were included. Maternal and neonatal microbiota profiles were obtained at birth by 16S rRNA gene profiling. Clinical records were collected. Geographical location influenced maternal-neonatal microbiota. Indeed, neonatal and maternal cores composed by nine genera each one were found independently of location. Geographical location was the most important variable that impact the overall structure of maternal and neoantal microbiota. For neonates, delivery mode effect on neonatal microbial community could modulate how the other perinatal factors, as geographical location or maternal BMI, impact the neoantal initial seeding. Furthermore, lower maternal pre-pregnancy BMI was associated with higher abundance of Faecalibacterium in maternal microbiota and members from Lachnospiraceae family in both mothers and infants. At genus-level, Chinese maternal-neonate dyads possessed higher number of phylogenetic shared microbiota than that of Spanish dyads. Bifidobacterium and Escherichia/Shigella were the genera most shared between dyads in the two groups highlighting their importance in neonatal colonization and mother-infant transmission. Our data showed that early gut microbiota establishment and development is affected by interaction of complex variables, where environment would be a critical factor.
Collapse
Affiliation(s)
- Yue Cheng
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Xin Cao
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Marta Calatayud
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Qi Qi
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Jing Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lingxia Zeng
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Izaskun Garcia-Mantrana
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Bei Han
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
28
|
McGill S, Burchmore RJS, Pomeroy PP, Kennedy MW. Is a Little Enough? Paucity of Immune Proteins in Serum of Precocial Neonates of a Marine Carnivoran—the Atlantic Grey Seal. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2021.802510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mammalian mothers usually provide their offspring with large quantities of immunoglobulins (antibodies) for circulation in blood, either trans-placentally before birth, via colostrum briefly thereafter, or, less commonly, from milk. Neonates of true, phocid seals, however, are peculiarly impoverished in serum immunoglobulins, the levels of which slowly increase but do not reach adult levels by the time of weaning. We investigated whether grey seal (Halichoerus grypus) neonates compensate through an elevation or rapid maturation in levels of serum innate immune factors, namely acute phase and complement proteins. Instead, their sera contained remarkably low levels of acute phase proteins (including C-reactive protein, haptoglobin, hemopexin, ceruloplasmin, orosomucoid), compared to their mothers, that barely increased to adult levels by weaning. For complement, there was a strong demarcation between the early activation and amplification cascade components (present at normal adult levels in pups) and the late lytic membrane attack complex and regulatory proteins (consistently at low relative levels). Phocid neonates therefore differ dramatically from land Carnivorans, such as dogs and cats, in early life immune protection. That neonatal phocids survive this apparent vulnerability to infections between birth and weaning prompts questions as to what other mechanisms protect them, and the adaptive value of their seeming vulnerability.
Collapse
|
29
|
Latino C, Gianatti EJ, Mehta S, Lo J, Devine A, Christophersen C. Does a high dietary intake of resistant starch affect glycaemic control and alter the gut microbiome in women with gestational diabetes? A randomised control trial protocol. BMC Pregnancy Childbirth 2022; 22:46. [PMID: 35042457 PMCID: PMC8764780 DOI: 10.1186/s12884-021-04366-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gestational Diabetes Mellitus (GDM) is prevalent with lasting health implications for the mother and offspring. Medical nutrition therapy is the foundation of GDM management yet achieving optimal glycaemic control often requires treatment with medications, like insulin. New dietary strategies to improve GDM management and outcomes are required. Gut dysbiosis is a feature of GDM pregnancies, therefore, dietary manipulation of the gut microbiota may offer a new avenue for management. Resistant starch is a fermentable dietary fibre known to alter the gut microbiota and enhance production of short-chain fatty acids. Evidence suggests that short-chain fatty acids improve glycaemia via multiple mechanisms, however, this has not been evaluated in GDM. METHODS An open-label, parallel-group design study will investigate whether a high dietary resistant starch intake or resistant starch supplement improves glycaemic control and changes the gut microbiome compared with standard dietary advice in women with newly diagnosed GDM. Ninety women will be randomised to one of three groups - standard dietary treatment for GDM (Control), a high resistant starch diet or a high resistant starch diet plus a 16 g resistant starch supplement. Measurements taken at Baseline (24 to 30-weeks' gestation), Day 10 and Day 56 (approximately 36 weeks' gestation) will include fasting plasma glucose levels, microbial composition and short-chain fatty acid concentrations in stool, 3-day dietary intake records and bowel symptoms questionnaires. One-week post-natal data collection will include microbial composition and short-chain fatty acid concentrations of maternal and neonatal stools, microbial composition of breastmilk, birthweight, maternal and neonatal outcomes. Mixed model analysis of variance will assess change in glycaemia and permutation-based multivariate analysis of variance will assess changes in microbial composition within and between intervention groups. Distance-based linear modelling will identify correlation between change in stool microbiota, short-chain fatty acids and measures of glycaemia. DISCUSSION To improve outcomes for GDM dyads, evaluation of a high dietary intake of resistant starch to improve glycaemia through the gut microbiome needs to be established. This will expand the dietary interventions available to manage GDM without medication. TRIAL REGISTRATION Australian New Zealand Clinical Trial Registry, ACTRN12620000968976p . Registered 28 September 2020.
Collapse
Affiliation(s)
- Cathy Latino
- School of Medical & Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
- Institute for Nutrition Research, Edith Cowan University, Joondalup, Western Australia, Australia.
- Department of Dietetics, Fiona Stanley Hospital, South Metropolitan Health Service, 11 Robin Warren Drive, Murdoch, 6150, Western Australia, Australia.
| | - Emily J Gianatti
- Department of Endocrinology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Shailender Mehta
- Department of Neonatology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
- School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Johnny Lo
- School of Science, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Amanda Devine
- School of Medical & Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Institute for Nutrition Research, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Claus Christophersen
- School of Medical & Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Integrative Metabolomics and Computational Biology, Edith Cowan University, Joondalup, Western Australia, Australia
- WA Human Microbiome Collaboration Centre - TrEnD Lab, School of Molecular & Life Sciences, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
30
|
Hill L, Sharma R, Hart L, Popov J, Moshkovich M, Pai N. The neonatal microbiome in utero and beyond: perinatal influences and long-term impacts. J LAB MED 2021. [DOI: 10.1515/labmed-2021-0131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
The neonatal microbiome offers a valuable model for studying the origins of human health and disease. As the field of metagenomics expands, we also increase our understanding of early life influences on its development. In this review we will describe common techniques used to define and measure the microbiome. We will review in utero influences, normal perinatal development, and known risk factors for abnormal neonatal microbiome development. Finally, we will summarize current evidence that links early life microbial impacts on the development of chronic inflammatory diseases, obesity, and atopy.
Collapse
Affiliation(s)
- Lee Hill
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Department of Human Biology, Division of Exercise Science and Sports Medicine , University of Cape Town , Cape Town , South Africa
| | - Ruchika Sharma
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- McMaster University , Hamilton , Canada
| | - Lara Hart
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
| | - Jelena Popov
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- University College Cork, College of Medicine and Health , Cork , Ireland
| | - Michal Moshkovich
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Faculty of Health Sciences , McMaster University , Hamilton , Canada
| | - Nikhil Pai
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition , McMaster Children’s Hospital, McMaster University , Hamilton , Canada
- Farncombe Family Digestive Health Research Institute , McMaster University , Hamilton , Canada
| |
Collapse
|
31
|
Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. Breast Milk: A Source of Functional Compounds with Potential Application in Nutrition and Therapy. Nutrients 2021; 13:1026. [PMID: 33810073 PMCID: PMC8005182 DOI: 10.3390/nu13031026] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is an unbeatable food that covers all the nutritional requirements of an infant in its different stages of growth up to six months after birth. In addition, breastfeeding benefits both maternal and child health. Increasing knowledge has been acquired regarding the composition of breast milk. Epidemiological studies and epigenetics allow us to understand the possible lifelong effects of breastfeeding. In this review we have compiled some of the components with clear functional activity that are present in human milk and the processes through which they promote infant development and maturation as well as modulate immunity. Milk fat globule membrane, proteins, oligosaccharides, growth factors, milk exosomes, or microorganisms are functional components to use in infant formulas, any other food products, nutritional supplements, nutraceuticals, or even for the development of new clinical therapies. The clinical evaluation of these compounds and their commercial exploitation are limited by the difficulty of isolating and producing them on an adequate scale. In this work we focus on the compounds produced using milk components from other species such as bovine, transgenic cattle capable of expressing components of human breast milk or microbial culture engineering.
Collapse
Affiliation(s)
- Cristina Sánchez
- Pharmacy Faculty, San Pablo-CEU University, 28003 Madrid, Spain;
| | - Luis Franco
- Medicine Faculty, Santiago de Compostela University, 15782 Santiago de Compostela, Spain;
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| |
Collapse
|
32
|
Hajiagha MN, Taghizadeh S, Asgharzadeh M, Dao S, Ganbarov K, Köse Ş, Kafil HS. Gut microbiota And Human Body Interactions; Its Impact on Health: a review. Curr Pharm Biotechnol 2021; 23:4-14. [PMID: 33397232 DOI: 10.2174/1389201022666210104115836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
Gut microbiota (GM) as an organ of the human body has a particular and autonomous function that related to it. This review aimed to investigate human intestinal and gut microbiota interaction and its impact on health. As a creation referable database about this dynamic and complex organ, several comprehensive projects are implemented by using culture-dependent (culturomics), culture independent methods (e.g metagenomics, mathematics model), and Gnotobiological together. This study was done by searching PubMed, Scopus and Google scholar database in the gut, health microbiota and interaction keywords. The first acquired microbiota during pregnancy or childbirth is colonized in the gut by using specific and non-specific mechanisms. That`s structure and shape reach relative stability with selection pressure along with host development until adulthood and keep its resilience against external or internal variables depending on the host genetics and negative feedback. Due to several research individuals have 2 functional group microbiota including the core (common between vast majorities human) and flexible (transient population) microbiome. The most important role of the GM in the human body can be summarized in three basic landscapes: metabolic, immune system, and gut-brain axis interaction. So that loss of microbial population balance will lead to disorder and disease.
Collapse
Affiliation(s)
| | - Sepehr Taghizadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mohammad Asgharzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Sounkalo Dao
- Faculté de Médecine, de Pharmacie et d'Odonto-Stomatologie (FMPOS), University of Bamako, Bamako. Mali
| | | | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir. Turkey
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
33
|
He Y, Peng X, Liu Y, Wu Q, Zhou Q, Hu L, Fang Z, Lin Y, Xu S, Feng B, Li J, Zhuo Y, Wu D, Che L. Effects of Maternal Fiber Intake on Intestinal Morphology, Bacterial Profile and Proteome of Newborns Using Pig as Model. Nutrients 2020; 13:E42. [PMID: 33375592 PMCID: PMC7823571 DOI: 10.3390/nu13010042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Dietary fiber intake during pregnancy may improve offspring intestinal development. The aim of this study was to evaluate the effect of maternal high fiber intake during late gestation on intestinal morphology, microbiota, and intestinal proteome of newborn piglets. Sixteen sows were randomly allocated into two groups receiving the control diet (CD) and high-fiber diet (HFD) from day 90 of gestation to farrowing. Newborn piglets were selected from each litter, named as CON and Fiber group, respectively. Maternal high fiber intake did not markedly improve the birth weight, but increased the body length, the ileal crypt depth and colonic acetate level. In addition, maternal high fiber intake increased the -diversity indices (Observed species, Simpson, and ACE), and the abundance of Acidobacteria and Bacteroidetes at phylum level, significantly increased the abundance of Bradyrhizobium and Phyllobacterium at genus level in the colon of newborn piglets. Moreover, maternal high fiber intake markedly altered the ileal proteome, increasing the abundances of proteins associated with oxidative status, energy metabolism, and immune and inflammatory responses, and decreasing abundances of proteins related to cellular apoptosis, cell structure, and motility. These findings indicated that maternal high fiber intake could alter intestinal morphology, along with the altered intestinal microbiota composition and proteome of offspring.
Collapse
Affiliation(s)
- Ying He
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Xie Peng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Yang Liu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Qing Wu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Qiang Zhou
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Liang Hu
- College of Food Science, Sichuan Agricultural University, Ya’an 625014, China;
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Jian Li
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - De Wu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.H.); (X.P.); (Y.L.); (Q.W.); (Q.Z.); (Z.F.); (Y.L.); (S.X.); (B.F.); (J.L.); (Y.Z.); (D.W.)
| |
Collapse
|
34
|
Hanson P, Weickert MO, Barber TM. Obesity: novel and unusual predisposing factors. Ther Adv Endocrinol Metab 2020; 11:2042018820922018. [PMID: 32489583 PMCID: PMC7238298 DOI: 10.1177/2042018820922018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/05/2020] [Indexed: 12/28/2022] Open
Abstract
To tackle the complexity of the global obesity epidemic, it is important to consider the many predisposing factors that underlie progressive and sustained weight gain. Some of the biological drivers for weight gain following initial weight loss include persistent changes in appetite hormones [including ghrelin and postprandial plasma peptide YY (PYY)], and 'persistent metabolic adaptation'. However, many factors within our busy, stressful modern-day environment seem to conspire towards promotion of weight gain. These include the effects of sleep deprivation on appetite regulation, and the effects of modern-day technology on 'attention competition'. These factors, combined with cultural and societal factors can result in a 'mindless' attitude regarding eating-related behaviour that is likely to predispose to weight gain. In addition to the external environment, our internal environment within the gut has also changed radically within the last few decades, resulting from changes in fibre intake, and increased ingestion of highly refined, sterilised and processed foods. Although contentious, these dietary changes have implications for our gut microbiota, and possible downstream effects on control of appetite and metabolism. In this brief review, we consider some of the novel predisposing factors for weight gain within our modern-day 21st century environments (both external and internal), and explore how legal terminology can help to conceptualise the numerous factors that contribute towards weight gain, and, ultimately the global obesity epidemic.
Collapse
Affiliation(s)
- Petra Hanson
- Clinical Sciences Research Laboratories, Warwick
Medical School, University Hospitals Coventry and Warwickshire, Clifford
Bridge Road, Coventry, CV2 2DX
- Warwickshire Institute for the Study of
Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and
Warwickshire, Clifford Bridge Road, Coventry, CV2 2DX
| | - Martin O. Weickert
- Clinical Sciences Research Laboratories, Warwick
Medical School, University Hospitals Coventry and Warwickshire, Clifford
Bridge Road, Coventry, CV2 2DX
- Warwickshire Institute for the Study of
Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and
Warwickshire, Clifford Bridge Road, Coventry, CV2 2DX
- Centre of Applied Biological & Exercise
Sciences (ABES), Faculty of Health & Life Sciences, Coventry University,
Coventry, UK
| | | |
Collapse
|
35
|
Maes M, Anderson G, Betancort Medina SR, Seo M, Ojala JO. Integrating Autism Spectrum Disorder Pathophysiology: Mitochondria, Vitamin A, CD38, Oxytocin, Serotonin and Melatonergic Alterations in the Placenta and Gut. Curr Pharm Des 2019; 25:4405-4420. [PMID: 31682209 DOI: 10.2174/1381612825666191102165459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND A diverse array of data has been associated with autism spectrum disorder (ASD), reflecting the complexity of its pathophysiology as well as its heterogeneity. Two important hubs have emerged, the placenta/prenatal period and the postnatal gut, with alterations in mitochondria functioning crucial in both. METHODS Factors acting to regulate mitochondria functioning in ASD across development are reviewed in this article. RESULTS Decreased vitamin A, and its retinoic acid metabolites, lead to a decrease in CD38 and associated changes that underpin a wide array of data on the biological underpinnings of ASD, including decreased oxytocin, with relevance both prenatally and in the gut. Decreased sirtuins, poly-ADP ribose polymerase-driven decreases in nicotinamide adenine dinucleotide (NAD+), hyperserotonemia, decreased monoamine oxidase, alterations in 14-3-3 proteins, microRNA alterations, dysregulated aryl hydrocarbon receptor activity, suboptimal mitochondria functioning, and decreases in the melatonergic pathways are intimately linked to this. Many of the above processes may be modulating, or mediated by, alterations in mitochondria functioning. Other bodies of data associated with ASD may also be incorporated within these basic processes, including how ASD risk factors such as maternal obesity and preeclampsia, as well as more general prenatal stressors, modulate the likelihood of offspring ASD. CONCLUSION Such a mitochondria-focussed integrated model of the pathophysiology of ASD has important preventative and treatment implications.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | | - Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Johanna O Ojala
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|