1
|
Gurunthalingam MP, Singh MP, Gaikwad NR. Biannual azithromycin mass drug administration for reduction of childhood mortality: a systematic review and meta-analysis. J Antimicrob Chemother 2025; 80:1197-1206. [PMID: 40129224 DOI: 10.1093/jac/dkaf092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Biannual mass drug administration of azithromycin (MDA-azithromycin) has been proposed as a strategy to reduce childhood mortality in high-mortality regions, particularly sub-Saharan Africa. However, its effectiveness across different age groups and potential risks, including antibiotic resistance, require further evaluation. METHODS We systematically searched PubMed, Cochrane CENTRAL, Web of Science and ClinicalTrials.gov through September 2024 for randomized controlled trials (RCTs) comparing biannual MDA-azithromycin to placebo in children aged 1-59 months. The primary outcomes were mortality in children <1 year and 12-59 months. Secondary outcomes included adverse events and antibiotic resistance. Data were analysed using a random-effects model in Review Manager 5.4, with heterogeneity assessed via I2. Trial sequential analysis (TSA) evaluated cumulative evidence reliability, and the Cochrane RoB2 tool assessed risk of bias. PROSPERO registration: CRD42024589170. RESULTS Five RCTs (691 235 children) were included. Among children <1 year, azithromycin showed a non-significant mortality reduction (RR: 0.90 [0.78, 1.04]; P = 0.14; I2 = 55%), with TSA indicating inconclusive evidence. Among children 12-59 months, MDA-azithromycin significantly reduced mortality (RR: 0.85 [0.79, 0.91]; P < 0.00001; I2 = 26%), with TSA confirming sufficient evidence. Adverse events were rare, but antibiotic resistance data were limited, warranting further monitoring. Evidence quality ranged from moderate to very low, with one trial at high risk of bias. CONCLUSION Biannual MDA-azithromycin significantly reduces mortality in children 12-59 months, supporting its use in high-mortality settings per WHO recommendations. Its impact on infants remains uncertain. Adverse events were minimal, but continued resistance surveillance is essential.
Collapse
Affiliation(s)
| | - Madhusudan Prasad Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Nitin Rewaram Gaikwad
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| |
Collapse
|
2
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
3
|
Alazwari A, Tafakori L, Johnstone A, Abdollahian M. Modeling the number of new cases of childhood type 1 diabetes using Poisson regression and machine learning methods; a case study in Saudi Arabia. PLoS One 2025; 20:e0321480. [PMID: 40279367 PMCID: PMC12027261 DOI: 10.1371/journal.pone.0321480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/06/2025] [Indexed: 04/27/2025] Open
Abstract
Diabetes mellitus stands out as one of the most prevalent chronic conditions affecting pediatric populations. The escalating incidence of childhood type 1 diabetes (T1D) globally is a matter of increasing concern. Developing an effective model that leverages Key Performance Indicators (KPIs) to understand the incidence of T1D in children would significantly assist medical practitioners in devising targeted monitoring strategies. This study models the number of monthly new cases of T1D and its associated KPIs among children aged 0 to 14 in Saudi Arabia. The study involved collecting de-identified data (n=377) from diagnoses made between 2010 and 2020, sourced from pediatric diabetes centers in three cities across Saudi Arabia. Poisson regression (PR), and various machine learning (ML) techniques, including random forest (RF), support vector machine (SVM), and K-nearest neighbor (KNN), were employed to model the monthly number of new T1D cases using the local data. The performance of these models was assessed using both numbers of KPIs and metrics such as the coefficient of determination ([Formula: see text]), root mean squared error (RMSE), and mean absolute error (MAE). Among various Poisson and ML models, both model considering birth weight over 3.5 kg, maternal age over 25 years at the child's birth, family history of T1D, and nutrition history, specifically early introduction to cow milk and model taking into account birth weight over 3.5 kg, maternal age over 25 years at the child's birth, and nutrition history (early introduction to cow milk) emerged as the best-reduced models. They achieved [Formula: see text] of (0.89,0.88), RMSE (0.82, 0.95) and MAE(0.62,0.67). Additionally, models with fewer KPIs, like model that considers maternal age over 25 years and early introduction to cow milk, achieved consistently high [Formula: see text] values ranging from 0.80 to 0.83 across all models. Notably, this model demonstrated smaller values of RMSE (0.92) and MAE (0.67) in the KNN model. Simplified models facilitate the efficient creation and monitoring of KPIs profiles. The findings can assist healthcare providers in collecting and monitoring influential KPIs, enabling the development of targeted strategies to potentially reduce, or reverse, the increasing incidence rate of childhood T1D in Saudi Arabia.
Collapse
Affiliation(s)
- Ahood Alazwari
- School of Science, RMIT University, Melbourne, Victoria, Australia
- School of Science, Al-Baha University, Al-Baha, Saudi Arabia
| | - Laleh Tafakori
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Alice Johnstone
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Mali Abdollahian
- School of Science, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Rai AK, Yadav M, Duary RK, Shukla P. Gut Microbiota Modulation Through Dietary Approaches Targeting Better Health During Metabolic Disorders. Mol Nutr Food Res 2025:e70033. [PMID: 40195821 DOI: 10.1002/mnfr.70033] [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: 10/05/2024] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
The impact of gut microbiota is known to play a significant role in an individual's metabolism and health. Many harmful food products or dietary imbalance adversely affect human health and changing lifestyle, environmental factors, and food habits may have their effect on gut microbiota. It has emerged that gut microbiota is regarded as an emerging metabolic organ, which is dependent on individual's diet and its composition. This review discusses the significance of lactic acid bacteria as a prominent inhabitant in the gut microbiota and the role of probiotics, prebiotics, and polyphenols to improve human health and metabolism. The role of fermented foods as an important source of probiotics and bioactive molecules is also discussed along with the role of gut microbiota in metabolic disorders like dyslipidemia, obesity, hypercholesterolemia, cancer, and hypertension. Finally, the review gives insights into the effective therapeutic prospects through gut microbiota alterations to tackle these metabolic disorders.
Collapse
Affiliation(s)
- Amit Kumar Rai
- BRIC-National Agri-Food and Biomanufacturing Institute (BRIC-NABI), SAS Nagar, Mohali, India
| | | | - Raj Kumar Duary
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
5
|
Chhibba T, Gros B, King JA, Windsor JW, Gorospe J, Leibovitzh H, Xue M, Turpin W, Croitoru K, Ananthakrishnan AN, Gearry RB, Kaplan GG. Environmental risk factors of inflammatory bowel disease: toward a strategy of preventative health. J Crohns Colitis 2025; 19:jjaf042. [PMID: 40065502 PMCID: PMC12010164 DOI: 10.1093/ecco-jcc/jjaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) involves a complex interplay between genetic, environmental, and microbial factors. Many of these environmental determinants are modifiable, offering opportunities to prevent disease or delay its onset. Advances in the study of preclinical IBD cohorts offer the potential to identify biomarkers that predict individuals at high risk of developing IBD, enabling targeted environmental interventions aimed at reducing IBD incidence. This review summarizes findings from 79 meta-analyses on modifiable environmental factors associated with the development of IBD. Identified risk factors include smoking, Western diets, ultra-processed foods, and early life antibiotic use, while protective factors include breastfeeding, Mediterranean diets rich in fiber, plant-based foods, and fish, along with an active physical lifestyle. Despite the promise shown by observational data, interventional or randomized controlled studies evaluating the efficacy of modifying environmental risk factors remain limited and mostly focus on dietary intervention. This review aims to inform the design of higher quality interventional and randomized controlled studies for disease prevention while providing actionable guidance to healthcare providers on reducing the risk of developing IBD through environmental modifications.
Collapse
Affiliation(s)
- Tarun Chhibba
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Beatriz Gros
- Department of Gastroenterology and Hepatology, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Madrid, Spain
| | - James A King
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Joseph W Windsor
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Julia Gorospe
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Haim Leibovitzh
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mingyue Xue
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Williams Turpin
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Richard B Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Gilaad G Kaplan
- Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
Corrias MV, Di Marco E, Bonaretti C, Squillario M, Amoroso L, Conte M, Ponzoni M, Biassoni R. In Infants with Neuroblastoma Standard Therapy Only Partially Reverts the Fecal Microbiome Dysbiosis Present at Diagnosis. Microorganisms 2025; 13:691. [PMID: 40142583 PMCID: PMC11946756 DOI: 10.3390/microorganisms13030691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
The fecal microbiomes of 15 infants with neuroblastoma (NB) at the onset of the disease and after standard-of-care therapy have been prospectively analyzed compared to those of age-matched healthy infants. By applying several algorithms to 16S sequencing, we found that the fecal microbiomes of infants with NB at onset were abundant in Pseudomonadota, including different descendants of Gammaproteobacteria. After completing therapy, their abundance decreased to a level like that observed in healthy infants. In contrast, the Bacillota that showed at the onset an abundance like that of healthy infants doubled their amount after treatment. In infants with NB, the beta diversity of the fecal microbiomes was significantly reduced compared to healthy infants and patients at the end of treatment. The Random Forest algorithm and the Reingold-Tilford heat tree showed that Enterobacteriaceae had a higher abundance at the onset, which declined after therapy. Picrust2 inferred pathway analysis indicated that the drug treatment was associated with a reduction in the polyamine pathway, highly represented in samples of NB at the onset. In conclusion, the dysbiosis observed in infants with NB at onset changed following standard-of-care treatment. Still, the composition at the end of treatment did not completely resemble that of healthy infants.
Collapse
Affiliation(s)
- Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Eddi Di Marco
- Central Laboratory, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy;
| | - Carola Bonaretti
- Molecular Diagnostics, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (C.B.); (R.B.)
| | | | - Loredana Amoroso
- Oncology Unit, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (L.A.); (M.C.)
| | - Massimo Conte
- Oncology Unit, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (L.A.); (M.C.)
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Roberto Biassoni
- Molecular Diagnostics, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (C.B.); (R.B.)
| |
Collapse
|
7
|
O’Neill L, Vasiloglou MF, Salesse F, Bailey R, Nogueira-de-Almeida CA, Al Dhaheri A, Cheikh Ismail L, Hwalla N, Mak TN. Impact of Fortified Whole Grain Infant Cereal on the Nutrient Density of the Diet in Brazil, the UAE, and the USA: A Dietary Modeling Study. CHILDREN (BASEL, SWITZERLAND) 2025; 12:384. [PMID: 40150666 PMCID: PMC11941295 DOI: 10.3390/children12030384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND/OBJECTIVES Complementary feeding (CF) influences future health outcomes. The aim of this study was to evaluate the impact of fortified whole grain infant cereal (WGIC), a complementary food, among 6- to 12-month-old infants on the nutrient density of the diet in three diverse settings: Brazil, the United Arab Emirates (UAE), and the US. METHODS Data from the Feeding Infants and Toddler Study (FITS), a collection of dietary intake studies based on 24-h-dietary recalls, from said countries was utilized. Nutrient intakes were calculated for infant cereal (IC) consumers and non-consumers. Diet modeling was applied to IC consumers to substitute their regular fortified IC with WGIC with improved fortifications. The study estimated the average nutrient density, mean adequacy ratio (MAR), and percentage inadequacy of the diet in both IC consumers and non-consumers. RESULTS The analyses indicated that infants who consumed IC had higher intakes of calcium, zinc, magnesium, iron, and vitamin D in the three countries. Reduced micronutrient inadequacies were observed among IC consumers, particularly in Brazil and the U.S. Diet modeling with WGIC revealed a significantly higher density of choline, magnesium, zinc, iron, fiber, and protein, as well as reduced inadequacies. The MAR was significantly improved in the three countries. CONCLUSIONS This study underscores the potential of fortified WGIC in increasing the nutrient density of the complementary diet. The intrinsic nutrients in whole grain infant cereals (WGICs) significantly enhance the nutrient density of the complementary diet. Given that whole grains play a role in preventing childhood obesity, their inclusion during CF may be crucial.
Collapse
Affiliation(s)
- Lynda O’Neill
- Nutrition Sciences, Nestlé Institute of Health Science, Nestlé Research, 1000 Lausanne, Switzerland;
| | - Maria F. Vasiloglou
- Nutrition Sciences, Nestlé Institute of Health Science, Nestlé Research, 1000 Lausanne, Switzerland;
| | - Fanny Salesse
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Regan Bailey
- Institute for Advancing Health through Agriculture, Texas A&M University, College Station, TX 77840, USA;
| | | | - Ayesha Al Dhaheri
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Leila Cheikh Ismail
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford OX1 2JD, UK
| | - Nahla Hwalla
- Department of Nutrition and Applied Sciences, American University of Beirut, Beirut P.O. Box 11-10236, Lebanon;
| | - Tsz Ning Mak
- Nestlé Institute of Health Science, 21 Biopolis Road, Singapore 618802, Singapore;
| |
Collapse
|
8
|
Berdowska I, Matusiewicz M, Fecka I. A Comprehensive Review of Metabolic Dysfunction-Associated Steatotic Liver Disease: Its Mechanistic Development Focusing on Methylglyoxal and Counterbalancing Treatment Strategies. Int J Mol Sci 2025; 26:2394. [PMID: 40141037 PMCID: PMC11942149 DOI: 10.3390/ijms26062394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial disorder characterized by excessive lipid accumulation in the liver which dysregulates the organ's function. The key contributor to MASLD development is insulin resistance (IR) which affects many organs (including adipose tissue, skeletal muscles, and the liver), whereas the molecular background is associated with oxidative, nitrosative, and carbonyl stress. Among molecules responsible for carbonyl stress effects, methylglyoxal (MGO) seems to play a major pathological function. MGO-a by-product of glycolysis, fructolysis, and lipolysis (from glycerol and fatty acids-derived ketone bodies)-is implicated in hyperglycemia, hyperlipidemia, obesity, type 2 diabetes, hypertension, and cardiovascular diseases. Its causative effect in the stimulation of prooxidative and proinflammatory pathways has been well documented. Since metabolic dysregulation leading to these pathologies promotes MASLD, the role of MGO in MASLD is addressed in this review. Potential MGO participation in the mechanism of MASLD development is discussed in regard to its role in different signaling routes leading to pathological events accelerating the disorder. Moreover, treatment strategies including approved and potential therapies in MASLD are overviewed and discussed in this review. Among them, medications aimed at attenuating MGO-induced pathological processes are addressed.
Collapse
Affiliation(s)
- Izabela Berdowska
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Małgorzata Matusiewicz
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| |
Collapse
|
9
|
Pai VV, Sarath AP, Kerkar Z. Gut microbiome in dermatology - A narrative review. Indian J Dermatol Venereol Leprol 2025; 0:1-11. [PMID: 40357977 DOI: 10.25259/ijdvl_1094_2024] [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: 07/22/2024] [Accepted: 12/08/2024] [Indexed: 05/15/2025]
Abstract
The gut microbiome and human body have co-evolved in a synergistic host-microbial relationship. The ideal composition of human gut microbiota is an elusive concept, but every individual has a unique gut microbiota profile with regional differences. Newer diagnostic techniques have helped identify different bacteria and their roles in health and disease. The gut microbiome composition is affected by various factors like age, diet, immune system, environmental factors, exercise, and drugs. The microbiome has varied roles in metabolism, immune response, immune tolerance and antimicrobial protection. Diet plays an important role in maintaining the gut microbial diversity. Loss of homoeostasis in the microbiome results in dysbiosis. Dysbiosis plays a role in many dermatological diseases like atopic dermatitis, psoriasis, acne, rosacea, hidradenitis suppurativa, connective tissue disorders and many other systemic conditions like obesity, diabetes, neurological disease and malignancy. Reconstitution of the gut microbiome ecology in the form of bacteriotherapy with the reintegration of certain strains of microbiota has a beneficial role in many of these disorders.
Collapse
Affiliation(s)
| | | | - Zenia Kerkar
- Department of Dermatology, Goa Medical College, Bambolim, India
| |
Collapse
|
10
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2025; 51:264-284. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [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: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Borer KT. Relevance of Milk Composition to Human Longitudinal Growth from Infancy Through Puberty: Facts and Controversies. Nutrients 2025; 17:827. [PMID: 40077697 PMCID: PMC11901938 DOI: 10.3390/nu17050827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Milk is the principal nutrient of newborn humans and a diagnostic feature of the order Mammalia. Its release is elicited as a reflex by infant sucking under the control of the hormone oxytocin. While it is recognized that breast milk optimally promotes infant longitudinal growth and development, this review explores facts and controversies regarding the extent to which the milks of several dairy animals and infant formula milk (IF) approximate special properties and bioactivities of breast milk. It also provides evidence that early exposure to undernutrition during the very rapid fetal and early infancy growth predominantly and permanently stunts longitudinal growth trajectory in both animals and humans and is often followed in later life by obesity and metabolic dysfunction, and sometimes also by precocious timing of sexual maturation. There is a knowledge gap as to whether there may be additional critical periods of nutritional vulnerability in human development, which is characterized by a relatively prolonged period of slow childhood growth bracketed by the rapid fetal-neonatal and pubertal growth spurts. It is also unclear whether any quantitative differences in caloric intake and supply during neonatal period may influence developmental fatness programming. A further knowledge gap exists regarding the role of infant microbiome composition and development in the possible epigenetic programming of longitudinal growth or fatness in later life. Extending the research of early developmental programming to the entire period of human growth from conception to the end of puberty, examining infant caloric intake and supply as possible factors modulating the epigenetic programming in favor of obesity, and examining the role of infant gut microbiome in developing infant's capacity to process nutrients may provide a better understanding of the interaction between critical nutritional influences in the control of human longitudinal growth and later-life obesity.
Collapse
Affiliation(s)
- Katarina T Borer
- School of Kinesiology, The University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
12
|
Wang K, Hu Y, Wu Y, Xu J, Zhao Y, Yang J, Li X. The Therapeutic Potential of Gut-Microbiota-Derived Metabolite 4-Phenylbutyric Acid in Escherichia coli-Induced Colitis. Int J Mol Sci 2025; 26:1974. [PMID: 40076603 PMCID: PMC11901052 DOI: 10.3390/ijms26051974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 03/14/2025] Open
Abstract
Pathogenic Escherichia coli (E. coli) is a widely distributed pathogen that can cause varying degrees of zoonotic diseases, and infected animals often experience intestinal inflammation accompanied by diarrhea and dysbiosis. Previously, for the first time, we isolated Escherichia coli primarily of type B2 from a large-scale dairy farm in Yunnan, China. The 16s rRNA sequencing showed significant differences in the gut microbiota of calves infected with B2 E. coli, with higher abundance of harmful bacteria and lower abundance of beneficial bacteria compared with healthy calves. The metabolomics indicated that the concentrations of oxoadipic acid, 16-oxopalmitate, oerillyl alcohol, palmitoleic acid, and 4-phenylbutyrate (4-PBA) were significantly higher in the healthy group than in the infected group. The mouse model was established to assess the regulatory effect of 4-PBA on E. coli-induced colitis. Both oral administration of 4-PBA and fecal microbiota transplantation (FMT) had strong resistance to E. coli infection, improved survival rate and body weight, reduced intestinal tissue damage, decreased the levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), and restrained TLR4/MyD88/NF-κB pathway. Our study demonstrated that 4-PBA could relieve E. coli-induced colitis by improving gut microbiota structure and inhibiting the expression of pro-inflammatory cytokines through the TLR4/MyD88/NF-κB pathway. The present finding reveals the therapeutic potential of the gut-microbiota-derived metabolite 4-PBA for the treatment of colitis caused by E. coli.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Yang
- College of Veterinary Medicine, Yunnan Agricultural University, No. 452 Fengyuan Road, Panlong District, Kunming 650201, China; (K.W.); (Y.H.); (Y.W.); (J.X.); (Y.Z.)
| | - Xiaobing Li
- College of Veterinary Medicine, Yunnan Agricultural University, No. 452 Fengyuan Road, Panlong District, Kunming 650201, China; (K.W.); (Y.H.); (Y.W.); (J.X.); (Y.Z.)
| |
Collapse
|
13
|
Gong D, Gao Y, Shi R, Xu X, Yu M, Zhang S, Wang L, Dong Q. The gastric microbiome altered by A4GNT deficiency in mice. Front Microbiol 2025; 16:1541800. [PMID: 40012782 PMCID: PMC11861098 DOI: 10.3389/fmicb.2025.1541800] [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: 12/08/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025] Open
Abstract
Background Selective antimicrobial effects have been found for α1,4-linked N-acetylglucosamine residues at the terminus of O-glycans attached to a core protein of gastric gland mucin. A4gnt encodes α1,4-N-acetylglucosaminyl transferase, which is responsible for the biosynthesis of α1,4-linked N-acetylglucosamine. The impact of A4GNT on the establishment and homeostasis of the gastric microbiome remains to be clarified. The aim of this study was to characterize the gastric microbiome in mice deficient for the production of α1,4-linked N-acetylglucosamine. Methods The gastric microbiome within A4gnt -/- mice and wild-type mice was analyzed using high-throughput sequencing of bacterial 16S rRNA. Results In A4gnt -/- mice, which spontaneously develop gastric cancer, the community structure of the gastric microbiome was altered. The relative abundance of mutagenic Desulfovibrio and proinflammatory Prevotellamassilia in these mice was significantly increased, especially 4 weeks after birth. The co-occurrence network appeared to be much more complex. Functional prediction demonstrated considerable decreases in the relative frequencies of functions associated with polysaccharide metabolism and transportation. Conclusion The distinct profile in A4gnt -/- mice demonstrated a vital role of A4GNT in the establishment of the gastric microbiome. A dysbiotic gastric microbiome may contribute to the spontaneous development of gastric cancer in mice.
Collapse
Affiliation(s)
- Dawei Gong
- Department of Gastroenterology, The Forth People's Hospital of Jinan, Jinan, China
- Central Laboratories, The Affiliated Qingdao Municipal Hospital of Dalian Medical University, Qingdao, China
| | - Yuqiang Gao
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| | - Rui Shi
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaona Xu
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| | - Mengchao Yu
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| | - Shumin Zhang
- Department of Gastroenterology, The Forth People's Hospital of Jinan, Jinan, China
| | - Lili Wang
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| | - Quanjiang Dong
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
14
|
Dera N, Kosińska-Kaczyńska K, Żeber-Lubecka N, Brawura-Biskupski-Samaha R, Massalska D, Szymusik I, Dera K, Ciebiera M. Impact of Early-Life Microbiota on Immune System Development and Allergic Disorders. Biomedicines 2025; 13:121. [PMID: 39857705 PMCID: PMC11762082 DOI: 10.3390/biomedicines13010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: The shaping of the human intestinal microbiota starts during the intrauterine period and continues through the subsequent stages of extrauterine life. The microbiota plays a significant role in the predisposition and development of immune diseases, as well as various inflammatory processes. Importantly, the proper colonization of the fetal digestive system is influenced by maternal microbiota, the method of pregnancy completion and the further formation of the microbiota. In the subsequent stages of a child's life, breastfeeding, diet and the use of antibiotics influence the state of eubiosis, which determines proper growth and development from the neonatal period to adulthood. The literature data suggest that there is evidence to confirm that the intestinal microbiota of the infant plays an important role in regulating the immune response associated with the development of allergic diseases. However, the identification of specific bacterial species in relation to specific types of reactions in allergic diseases is the basic problem. Background: The main aim of the review was to demonstrate the influence of the microbiota of the mother, fetus and newborn on the functioning of the immune system in the context of allergies and asthma. Methods: We reviewed and thoroughly analyzed the content of over 1000 articles and abstracts between the beginning of June and the end of August 2024. Over 150 articles were selected for the detailed study. Results: The selection was based on the PubMed National Library of Medicine search engine, using selected keywords: "the impact of intestinal microbiota on the development of immune diseases and asthma", "intestinal microbiota and allergic diseases", "the impact of intrauterine microbiota on the development of asthma", "intrauterine microbiota and immune diseases", "intrauterine microbiota and atopic dermatitis", "intrauterine microbiota and food allergies", "maternal microbiota", "fetal microbiota" and "neonatal microbiota". The above relationships constituted the main criteria for including articles in the analysis. Conclusions: In the present review, we showed a relationship between the proper maternal microbiota and the normal functioning of the fetal and neonatal immune system. The state of eubiosis with an adequate amount and diversity of microbiota is essential in preventing the development of immune and allergic diseases. The way the microbiota is shaped, resulting from the health-promoting behavior of pregnant women, the rational conduct of the medical staff and the proper performance of the diagnostic and therapeutic process, is necessary to maintain the health of the mother and the child. Therefore, an appropriate lifestyle, rational antibiotic therapy as well as the way of completing the pregnancy are indispensable in the prevention of the above conditions. At the same time, considering the intestinal microbiota of the newborn in relation to the genera and phyla of bacteria that have a potentially protective effect, it is worth noting that the use of suitable probiotics and prebiotics seems to contribute to the protective effect.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, 02-781 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Diana Massalska
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Kacper Dera
- Pediatric Ward, Department of Pediatrics, Center of Postgraduate Medical Education, Bielański Hospital, 01-809 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| |
Collapse
|
15
|
van Beek N, Katavisto I, Lehto M, Kolho KL, de Vos WM, Salonen A, Korpela K. Host-microbiota interactions in the infant gut revealed by daily faecal sample time series. MICROBIOME RESEARCH REPORTS 2024; 4:13. [PMID: 40207273 PMCID: PMC11977378 DOI: 10.20517/mrr.2024.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 04/11/2025]
Abstract
Aim: This study aims to explore the interplay between host immune factors and gut microbiota in human infants in vivo using time-series daily stool samples and identify biomarkers of host-microbe interactions. Methods: 216 faecal samples collected from infants aged 5-6 or 11-12 months were analysed for gut microbiota composition, total bacterial load, and biomarkers of immune function. Results: We identified indications of microbial stimulation of eosinophil cationic protein (ECP), IgA, calprotectin (Cal), intestinal alkaline phosphatase (IAP), and Bactericidal/permeability-increasing protein (BPI) at 6 and 12 months, as well as stimulation of lipocalin 2 (LCN2), lactoferrin (LTF), and alpha-defensin-5 only at 6 months. The associations between biomarker concentrations and bacterial population growth were primarily positive at 6 months and mostly negative at 12 months, suggesting increasing host regulation of the microbiota with age. The exceptions were IAP, which was predictive of declining bacterial populations at both time points, and Cal, whose associations changed from negative at 6 months to positive at 12 months. Conclusion: There is an age-associated development in the correlation pattern between bacterial population growth and the biomarker concentrations, suggesting that host-microbe interactions change during early development. Albumin appeared as a potential marker of gut permeability, while LCN2 seemed to correlate with gut transit time. Mucin degradation appeared to decrease with age. Mucin2 and IAP emerged as potentially important regulators of the bacterial populations in the infant gut. The study demonstrates the utility of biomarker and bacteria profiling from daily stool samples for analysing in vivo associations between the immune system and the gut microbiota and provides evidence of host regulation of the microbiota in infants.
Collapse
Affiliation(s)
- Nienke van Beek
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Iiris Katavisto
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki 00250, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki 00014, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Kaija-Leena Kolho
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Faculty of Medicine, University of Helsinki and Children’s Hospital, Helsinki University Hospital HUS, Helsinki 00014, Finland
| | - Willem M. de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen 6700 EH, the Netherlands
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| |
Collapse
|
16
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Sabaredzovic A, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Philippat C, Eggesbø M, Lepage P, Slama R. Associations between pre- and post-natal exposure to phthalate and DINCH metabolites and gut microbiota in one-year old children. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125204. [PMID: 39490662 DOI: 10.1016/j.envpol.2024.125204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The gut microbiota is a collection of symbiotic microorganisms in the gastrointestinal tract. Its sensitivity to chemicals with widespread exposure, such as phthalates, is little known. We aimed to investigate the impact of perinatal exposure to phthalates on the infant gut microbiota at 12 months of age. Within SEPAGES cohort (Suivi de l'Exposition à la Pollution Atmosphérique durant la Grossesse et Effet sur la Santé), we assessed 13 phthalate metabolites and 2 di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH) metabolites in repeated urine samples collected in pregnant women and their offspring. We obtained stool samples from 356 children at 12 months of age and sequenced the V3-V4 region of the 16S rRNA gene, allowing gut bacterial profiling. We used single-chemical (linear regressions) and mixture (BKMR, Bayesian Kernel Machine Regression) models to examine associations of phthalates and DINCH metabolites, with gut microbiota indices of α-diversity (specific richness and Shannon diversity) and the relative abundances of the most abundant microbiota phyla and genera. After correction for multiple testing, di(2-ethylhexyl) phthalate (ΣDEHP), diethyl phthalate (DEP) and bis(2-propylheptyl) phthalate (DPHP) metabolites 12-month urinary concentrations were associated with higher Shannon α-diversity of the child gut microbiota in single-chemical models. The multiple-chemical model (BKMR) suggested higher α-diversity with exposure to the phthalate mixture at 12 months, driven by the same phthalates. There were no associations between phthalate and DINCH exposure biomarkers at other time points and α-diversity after correction for multiple testing. ΣDEHP metabolites concentration at 12 months was associated with higher Coprococcus genus. Finally, ΣDEHP exposure at 12 months tended to be associated with higher phylum Firmicutes, an association not maintained after correction for multiple testing. Infancy exposure to phthalate might disrupt children's gut microbiota. The observed associations were cross-sectional, so that reverse causality cannot be excluded.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France.
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Azemira Sabaredzovic
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Merete Eggesbø
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway; Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France; SMILE, Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France; PARSEC, Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France
| |
Collapse
|
17
|
Nikola L, Iva L. Gut microbiota as a modulator of type 1 diabetes: A molecular perspective. Life Sci 2024; 359:123187. [PMID: 39488260 DOI: 10.1016/j.lfs.2024.123187] [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: 05/22/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Type 1 diabetes (T1D) is defined as an autoimmune metabolic disorder, characterized by destruction of pancreatic β-cells and high blood sugar levels. If left untreated, T1D results in severe health complications, including cardiovascular and kidney disease, as well as nerve damage, with ultimately grave consequences. Besides the role of genetic and certain environmental factors in T1D development, in the last decade, one new player emerged to affect T1D pathology as well, and that is a gut microbiota. Dysbiosis of gut bacteria can contribute to T1D by gut barrier disruption and the activation of autoimmune response, leading to the destruction of insulin producing cells, causing the development and aggravation of T1D symptoms. The relationship between gut microbiota and diabetes is complex and varies between individuals and additional research is needed to fully understand the effects of gut microbiome alternations in T1D pathogenesis. Therefore, the goal of this review is to understand the current knowledge in underlying molecular mechanism of gut microbiota effects, which leads to the new approaches for further studies in the prevention and treatment of T1D.
Collapse
Affiliation(s)
- Lukic Nikola
- Laboratory for Molecular Biology and Endocrinology, Institute of Nuclear Sciences "Vinca", National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Lukic Iva
- Laboratory for Molecular Biology and Endocrinology, Institute of Nuclear Sciences "Vinca", National Institute of the Republic of Serbia, University of Belgrade, Serbia.
| |
Collapse
|
18
|
S S, L.S. D, Rajendran P, N H, Singh S A. Exploring the potential of probiotics in Alzheimer's disease and gut dysbiosis. IBRO Neurosci Rep 2024; 17:441-455. [PMID: 39629018 PMCID: PMC11612366 DOI: 10.1016/j.ibneur.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/06/2024] Open
Abstract
Alzheimer's disease is a fatal neurodegenerative disorder that causes memory loss and cognitive decline in older people. There is increasing evidence suggesting that gut microbiota alteration is a cause of Alzheimer's disease pathogenesis. This review explores the link between gut dysbiosis and the development of Alzheimer's disease contributing to neuroinflammation, amyloid β accumulation, and cognitive decline. We examine the recent studies that illustrate the gut-brain axis (GBA) as a bidirectional communication between the gut and brain and how its alteration can influence neurological health. Furthermore, we discuss the potential of probiotic supplementation as a management approach to restore gut microbiota balance, and ultimately improve cognitive function in AD patients. Based on current research findings, this review aims to provide insights into the promising role of probiotics in Alzheimer's disease management and the need for further investigation into microbiota-targeted interventions.
Collapse
Affiliation(s)
- Sowmiya S
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Dhivya L.S.
- Department of Pharmaceutical Chemistry, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Praveen Rajendran
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Harikrishnan N
- Department of pharmaceutical analysis, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Ankul Singh S
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| |
Collapse
|
19
|
Kapoor B, Biswas P, Gulati M, Rani P, Gupta R. Gut microbiome and Alzheimer's disease: What we know and what remains to be explored. Ageing Res Rev 2024; 102:102570. [PMID: 39486524 DOI: 10.1016/j.arr.2024.102570] [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: 06/18/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
With advancement in human microbiome research, an increasing number of scientific evidences have endorsed the key role of gut microbiota in the pathogenesis of Alzheimer disease. Microbiome dysbiosis, characterized by altered diversity and composition, as well as rise of pathobionts influence not only various gut disorder but also central nervous system disorders such as AD. On the basis of accumulated evidences of past few years now it is quite clear that the gut microbiota can control the functions of the central nervous system (CNS) through the gut-brain axis, which provides a new prospective into the interactions between the gut and brain. The main focus of this review is on the molecular mechanism of the crosstalk between the gut microbiota and the brain through the gut-brain axis, and on the onset and development of neurological disorders triggered by the dysbiosis of gut microbiota. Due to microbiota dysbiosis the permeability of the gut and blood brain barrier is increased which may mediate or affect AD. Along with this, bacterial population of the gut microbiota can secrete amyloid proteins and lipopolysaccharides in a large quantity which may create a disturbance in the signaling pathways and the formation of proinflammatory cytokines associated with the pathogenesis of AD. These topics are followed by a critical analysis of potential intervention strategies targeting gut microbiota dysbiosis, including the use of probiotics, prebiotics, metabolites, diets and fecal microbiota transplantation. The main purpose of this review includes the summarization and discussion on the recent finding that may explain the role of the gut microbiota in the development of AD. Understanding of these fundamental mechanisms may provide a new insight into the novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Pratim Biswas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia
| | - Pooja Rani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
20
|
Abenavoli L, Scarlata GG, Scarpellini E, Procopio AC, Ponziani FR, Boccuto L, Cetkovic N, Luzza F. Therapeutic success in primary biliary cholangitis and gut microbiota: a safe highway? Minerva Gastroenterol (Torino) 2024; 70:430-441. [PMID: 38240684 DOI: 10.23736/s2724-5985.23.03590-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Primary biliary cholangitis (PBC) is a chronic, cholestatic, autoimmune disease, characterized by destruction of bile ducts. PBC predominantly affects women between 40 and 60 years of age. The presence of antimitochondrial antibodies (AMA) is a serological feature of PBC. These highly specific antibodies are found in about 95% of patients with the disease. The family of enzymes located in the inner membrane of the mitochondria, called the 2-oxo-acid dehydrogenase complex represents the target of the AMA. Ursodeoxycholic acid (UDCA) is a synthetic bile acid capable of protecting cholangiocytes from cholestatic damage caused by the accumulation of bile acids with a mechanism of action not yet well clarified. UDCA represents the gold standard therapy for PBC patients with recommended dose of 13-15 mg/kg/day. However, not every patient responds to therapy. On the other hand, the gut microbiota plays a key role in the onset of PBC through still unclear biochemical pathways. Less is known about its role as a potential biomarker after drug treatment. Actually, few studies analyzed the changes in gut microbiota composition before and after UDCA treatment. For this reason, this review represents an examination of the studies carried out on changes in gut microbiota composition in patients affected by PBC before and after treatment.
Collapse
Affiliation(s)
- Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy -
| | - Giuseppe Gm Scarlata
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Emidio Scarpellini
- Department of Translational Research in Gastrointestinal Disorders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Leuven, Belgium
| | - Anna C Procopio
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Francesca R Ponziani
- Digestive Disease Center (C.E.M.A.D.), IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Luigi Boccuto
- School of Nursing, Clemson University, Clemson, SC, USA
| | - Nenad Cetkovic
- Department of Obstetrics and Gynecology, Clinical Center of Vojvodina, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Francesco Luzza
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
21
|
Badewy R, Glogauer M, Connor KL, Sgro M, Lai JY, Bazinet RP, Tenenbaum HC, Azarpazhooh A. The unrevealed links: periodontal health, human milk composition, and infant gut microbiome dynamics. Prim Health Care Res Dev 2024; 25:e62. [PMID: 39540631 DOI: 10.1017/s1463423624000215] [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] [Indexed: 11/16/2024] Open
Abstract
AIM This review aims to identify the mechanistic relationships related to periodontal diseases and its possible association with changes in human milk composition and the composition and function of infants' gut microbiome. BACKGROUND Maternal health conditions, especially inflammatory, are associated with altered human milk composition. It is not known whether maternal oral inflammatory diseases, including periodontal diseases, deleteriously affect human milk composition. METHODS A narrative review was conducted according to SANRA, the Scale for the Assessment of Narrative Review Articles, guidelines. PubMed, Google Scholar, and Cochrane database of systematic reviews were searched from September 2019 up to December 2023 using keywords such as breast/human milk, maternal health/infections, and periodontal diseases. Reference lists of relevant articles were also screened. Our primary outcome of interest was human milk composition (i.e., any changes in macronutrients, immunological components, etc.). Secondary outcomes included changes in human milk microbiome and subsequent changes in the infant gut microbiome. Outcomes were synthesized using a narrative approach where the existing evidence and current literature were summarized. No risk of bias assessment of the studies was performed in this review. FINDINGS The search yielded no studies investigating the relationship between periodontal diseases in nursing mothers and changes in human milk composition. However, a dose-response relationship exists between the severity of periodontal diseases and the risk of adverse pregnancy outcomes such as preterm birth. Mastitis and diabetes affected milk lipids. Immunoglobulin A (sIgA) was increased in mastitis, whereas reduced concentrations were reported in diabetes. Potential biological pathways through which periodontal diseases can negatively affect human milk composition include the systemic dissemination of inflammatory cytokines like IL-6, PGE2, and tumor necrosis factor (TNF)-β that can be up-regulated by bacterial by-products. This biological plausibility needs to be investigated, given the potentially negative impact on the quality of human milk that could be caused by periodontal inflammation.
Collapse
Affiliation(s)
- Rana Badewy
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dental Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Michael Sgro
- Department of Pediatrics, and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Pediatrics, Division of Neonatology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jim Yuan Lai
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Howard C Tenenbaum
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| | - Amir Azarpazhooh
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
22
|
Hasegawa-Ishii S, Komaki S, Asano H, Imai R, Osaki T. Chronic nasal inflammation early in life induces transient and long-term dysbiosis of gut microbiota in mice. Brain Behav Immun Health 2024; 41:100848. [PMID: 39280089 PMCID: PMC11402449 DOI: 10.1016/j.bbih.2024.100848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 09/18/2024] Open
Abstract
The gut microbiota begins to colonize the host body following birth, develops during the suckling period and changes to the adult type after weaning. The early gut microbiota during the suckling period is thought to have profound effects on the host physiology throughout life but it is still unclear whether early dysbiosis is retained lifelong. Our previous study indicated that chronic nasal inflammation induces dysbiosis of gut microbiota in adult mice. In the present study, we addressed the question as to whether early exposure to chronic nasal inflammation induces dysbiosis, and if so, whether the dysbiosis is retained until adulthood and the sex differences in this effect. Male and female mice received repeated intranasal administration of lipopolysaccharide (LPS) or saline twice a week from P7 to P24 and were weaned at P24. The cecal contents were obtained for 16S rRNA analysis at 2 time points: at 4 weeks (wks), just after weaning, and at maturation to adulthood at 10 wks. The body weight did not differ between saline- and LPS-treated mice till around weaning, suggesting that the mothers' milk was given similarly to all mice. At 4 wks, the beta diversity was significantly different between saline- and LPS-treated male and female mice and the composition of the gut microbiota changed in LPS-treated mice. The abundance of phylum Bacteroidota tended to decrease and that of Firmicutes increased in LPS-treated male mice, while the abundance of Deferribacterota increased in LPS-treated female mice. At 10 wks, the beta diversity was not different between saline- and LPS-treated mice, but the abundance of family Lachnospiraceae significantly decreased in LPS-treated male and female mice by LEfSe analysis. Together, chronic nasal inflammation early in life caused transient and long-term dysbiosis of gut microbiota, which may contribute to the onset and progress of metabolic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sanae Hasegawa-Ishii
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Suzuho Komaki
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Hinami Asano
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Ryuichi Imai
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, 181-8612, Japan
| | - Takako Osaki
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| |
Collapse
|
23
|
Long J, Zhang J, Zeng X, Wang M, Wang N. Prevention and Treatment of Alzheimer's Disease Via the Regulation of the Gut Microbiota With Traditional Chinese Medicine. CNS Neurosci Ther 2024; 30:e70101. [PMID: 39508315 PMCID: PMC11541599 DOI: 10.1111/cns.70101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD) is caused by a variety of factors, and one of the most important factors is gut microbiota dysbiosis. An imbalance in the gut mincrobiota have been shown to change the concentrations of lipopolysaccharide and short-chain fatty acids. These microorganisms synthesize substances that can influence the levels of a variety of metabolites and cause multiple diseases through the immune response, fatty acid metabolism, and amino acid metabolism pathways. Furthermore, these metabolic changes promote the formation of β-amyloid plaques and neurofibrillary tangles. Thus, the microbiota-gut-brain axis plays an important role in AD development. In addition to traditional therapeutic drugs such as donepezil and memantine, traditional Chinese medicines (TCMs) have also showed to significantly decrease the severity of AD symptoms and suppress the underlying related mechanisms. We searched for studies on the effects of different herbal monomers, single herbs, and polyherbal formulas on the gut microbiota of AD patients and identified the relevant pathways through which the gut microbiota affected AD. We conclude that improvements in the gut microbiota not only decrease the occurrence of inflammatory reactions but also reduce the deposition of central pathological products. Herbal monomers have a stronger effect on improving of central pathology. Polyherbal formulas have the most extensive effect on the gut microbiota in patients with AD. Among the effects of formulas, the anti-inflammatory effect is the most essential and is also the main concern regarding the use of TCMs in treating AD from the viewpoint of the gut microbiota. We hope that this review will be helpful for providing new ideas for the clinical application of TCMs in the treatment of AD.
Collapse
Affiliation(s)
- Jinyao Long
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jiani Zhang
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Xin Zeng
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Min Wang
- Dongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Ningqun Wang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
24
|
Li Y, Han Y, Wang X, Yang X, Ren D. Kiwifruit Polysaccharides Alleviate Ulcerative Colitis via Regulating Gut Microbiota-Dependent Tryptophan Metabolism and Promoting Colon Fucosylation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23859-23874. [PMID: 39432373 DOI: 10.1021/acs.jafc.4c06435] [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: 10/23/2024]
Abstract
A previous study showed that kiwifruit polysaccharide (KFP) has benefits in relieving intestinal inflammation, while the underlying mechanism remains unresolved. The objective of this study was to investigate the regulatory effect of KFP on the gut microbiota metabolism and intestinal barrier of ulcerative colitis (UC) mice induced by dextran sulfate sodium (DSS). KFP significantly improved the UC symptoms including weight loss, shortened colon length, splenomegaly, diarrhea, hematochezia, and colon inflammation of mice. In addition, KFP could alleviate DSS-caused gut microbiota dysbiosis and increase the levels of short-chain fatty acids in the cecal contents of mice. Furthermore, the results of nontargeted and targeted metabolomics analysis combined with antibiotic treatment revealed that KFP could regulate gut microbiota-dependent tryptophan metabolism, activate the aryl hydrocarbon receptor (AhR) in colon cells, and enhance interleukin-22 production and tight junction proteins' (ZO-1, occludin, and claudin3) expression to repair the intestinal barrier in UC mice. Immunofluorescence results showed that KFP significantly upregulated the conjunction of lectin WGA and UEA1 in the UC mouse colon, implying that KFP promoted fucosylation in the colon. These results suggest that KFP alleviates UC primarily via targeting the gut microbiota involved in the AhR pathway and upregulating colon fucosylation.
Collapse
Affiliation(s)
- Yixiao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yanhui Han
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xuejie Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
25
|
Okolie MC, Edo GI, Ainyanbhor IE, Jikah AN, Akpoghelie PO, Yousif E, Zainulabdeen K, Isoje EF, Igbuku UA, Orogu JO, Owheruo JO, Essaghah AEA, Umar H. Gut microbiota and immunity in health and diseases: a review. PROCEEDINGS OF THE INDIAN NATIONAL SCIENCE ACADEMY 2024. [DOI: 10.1007/s43538-024-00355-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/24/2024] [Indexed: 01/03/2025]
|
26
|
Mousavian AH, Zare Garizi F, Ghoreshi B, Ketabi S, Eslami S, Ejtahed HS, Qorbani M. The association of infant and mother gut microbiomes with development of allergic diseases in children: a systematic review. J Asthma 2024; 61:1121-1135. [PMID: 38506489 DOI: 10.1080/02770903.2024.2332921] [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: 12/20/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE It is believed that gut microbiota alteration leads to both intestinal and non-intestinal diseases in children. Since infants inherit maternal microbiota during pregnancy and lactation, recent studies suggest that changes in maternal microbiota can cause immune disorders as well. This systematic review was designed to assess the association between the child and mother's gut microbiome and allergy development in childhood. DATA SOURCES In this systematic review, international databases including PubMed, Scopus, and ISI/WOS were searched until January 2023 to identify relevant studies. STUDY SELECTIONS Observational studies that analyzed infant or maternal stool microbiome and their association with allergy development in children were included in this study. Data extraction and quality assessment of the included studies were independently conducted by two researchers. RESULTS Of the 1694 papers evaluated, 21 studies examined neonate gut microbiome by analyzing stool samples and six studies examined maternal gut microbiota. A total of 5319 participants were included in this study. Asthma followed by eczema and dermatitis were the most common allergy disorders among children. Urbanization caused a lack of diversity in the bacterial microbiota as well as lower levels of Bifidobacterium and Lachnospira associated with a higher risk of allergy. In contrast, higher levels of Roseburia and Flavonifractor were associated with lower allergy risk. CONCLUSIONS This systematic review shows that gut microbiota may be associated with allergy development. Further studies are required to provide a definitive answer.
Collapse
Affiliation(s)
- Amir-Hossein Mousavian
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Zare Garizi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Behnaz Ghoreshi
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Siavash Ketabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
27
|
Ma G, Li Y, Tye KD, Huang T, Tang X, Luo H, Wang D, Zhou J, Li Z, Xiao X. The effect of oral probiotics in the last trimester on the human milk and infant gut microbiotas at six months postpartum: A randomized controlled trial. Heliyon 2024; 10:e37157. [PMID: 39286230 PMCID: PMC11402683 DOI: 10.1016/j.heliyon.2024.e37157] [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: 02/06/2024] [Revised: 07/30/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Objective The main aim of this study was to evaluate the effect of oral probiotics on the human milk microbiota and determine whether that influenced infant microbiota development. Methods A total of 27 pregnant women were recruited; 14 were assigned to the probiotic group, and the rest were assigned to the control group. Their infants were likewise assigned to the probiotic group or the control group. Pregnant women in the probiotic group received probiotic supplementation from 32 weeks of gestation until delivery. Human milk samples and infant fecal samples were collected at 6 months after delivery, and 16S rRNA sequencing was used to analyze the composition of the human milk and infant gut microbiota (NCT06241222). Results In the control group, bacterial microbiota were detected in 8 out of 13 milk samples, whereas in the probiotic group, only 6 out of 14 milk samples contained bacterial microbiota. We examined the composition of the human milk and infant gut microbiota in both the control and probiotic groups. Spearman correlation analysis revealed that various genera in human milk were correlated with the infant gut microbiota. The Linear discriminant analysis effect size (LEfSe) showed that 6 bacteria in the human milk microbiota in the control group were significantly more abundant than those in the probiotic group. Nine bacteria were significantly more abundant in the human milk microbiota in the probiotic group than the control group. According to the LEfSe results, 11 bacteria in the infant gut microbiota in the control group were significantly more abundant than those in the probiotic group. Fourteen bacteria were significantly more abundant in the infant gut microbiota in the probiotic group than in the control group. Conclusion The infant gut microbiota at 6 months has a complicated relationship with the maternal human milk microbiota. Oral probiotic supplementation can change the composition of the human milk microbiota and the infant gut microbiota.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yimi Li
- Department of Obstetrics and Gynecology, Dangyang People's Hospital, Dangyang, China
| | - Kian Deng Tye
- Department of Obstetrics and Gynecology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongju Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhe Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
28
|
Singh K, Gupta PK, Kumar A, Singh BM. Characterization of Swarna Bhasma and Swarnaprash and Determination of Antimicrobial Properties Against Gut-Pathobiont and Symbiont. Biol Trace Elem Res 2024:10.1007/s12011-024-04353-8. [PMID: 39240307 DOI: 10.1007/s12011-024-04353-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
Ayurveda has a long-standing tradition of healthcare in Southeast Asia. Swarnaprash, a classical Ayurveda preparation, is commonly given as the pre-lacteal feed to neonates to prevent labor-related complications and infections. It comprises incinerated gold particles (InAuP/Swarna Bhasma), honey (Madhu), and clarified butter oil (CBO/Cow Ghrita). This in vitro study aimed to evaluate the therapeutic potential of the individual ingredients and combinations of Swarnaprash against selected neonatal gut pathobionts and symbionts. The study employed sophisticated instruments, including SEM with EDAX and X-ray diffraction analysis, to investigate the shape and structural disparities in the ingredients of Swarnaprash. The reported size of gold particles in Swarnaprash ranges from 0.6 to 9.5 µm. These particles are relatively smaller than those in Swarna Bhasma but larger than synthetic gold particles. Swarnaprash demonstrated both bactericidal and bacteriostatic activity against selected neonatal gut pathobionts, with the largest inhibition zones observed for P. aeruginosa and S. Typhi. It surpassed the individual efficacy of its components-Prash, InAuPs, honey, or CBO alone. Notably, Swarnaprash did not affect the selected beneficial gut bacteria. The results warrant further in vivo and clinical studies to explore the effects of Swarnaprash on neonatal gut flora, which would provide vital information for research in neonatal healthcare.
Collapse
Affiliation(s)
- Kavita Singh
- Department of Kaumarabhritya/Balroga, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Prashant Kumar Gupta
- Department of Kaumarabhritya, All India Institute of Ayurveda, Sarita Vihar, New Delhi, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, India.
| | - Brij Mohan Singh
- Department of Kaumarabhritya/Balroga, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
29
|
Kim AR, Jeon SG, Kim HR, Hong H, Yoon YW, Lee BM, Yoon CH, Choi SJ, Jang MH, Yang BG. Preventive and Therapeutic Effects of Lactiplantibacillus plantarum HD02 and MD159 through Mast Cell Degranulation Inhibition in Mouse Models of Atopic Dermatitis. Nutrients 2024; 16:3021. [PMID: 39275335 PMCID: PMC11396792 DOI: 10.3390/nu16173021] [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: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/16/2024] Open
Abstract
As the relationship between the gut microbiome and allergies becomes better understood, targeted strategies to prevent and treat allergies through gut microbiome modulation are being increasingly developed. In the study presented herein, we screened various probiotics for their ability to inhibit mast cell degranulation and identified Lactiplatibacillus plantarum HD02 and MD159 as effective candidates. The two strains significantly attenuated vascular permeability induced by mast cell degranulation in a passive cutaneous anaphylaxis (PCA) model and, in the MC903-induced murine atopic dermatitis (AD) model, demonstrated comparable preventive effects against allergies, reducing blood levels of MCPT-1 (mast cell protease-1) and total IgE. In the house dust mite (HDM)-induced murine AD model, both L. plantarum HD02 and MD159 showed therapeutic effects, with L. plantarum HD02 demonstrating superior efficacy. Nevertheless, L. plantarum MD159 better suppressed transepidermal water loss (TEWL). Furthermore, L. plantarum HD02 and MD159 significantly increased the number of splenic Foxp3+ regulatory T cells, with L. plantarum MD159 having a more pronounced effect. However, only L. plantarum HD02 achieved a reduction in immune cells in the draining lymph nodes. Our findings highlight L. plantarum HD02 and MD159 as promising candidates for the prevention and treatment of allergies, demonstrating significant efficacy in suppressing mast cell degranulation, reducing the number of allergy biomarkers, and modulating immune responses in experimental models of AD. Their distinct mechanisms of action suggest potential complementary roles in addressing allergic diseases, underscoring their therapeutic promise in clinical applications.
Collapse
Affiliation(s)
- A-Ram Kim
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Seong-Gak Jeon
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Hyung-Ran Kim
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Heeji Hong
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Yong Won Yoon
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Byung-Min Lee
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Chung Hoo Yoon
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Soo Jin Choi
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Myoung Ho Jang
- Research Institute, GI Innovation Inc., Seoul 05855, Republic of Korea
| | - Bo-Gie Yang
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| |
Collapse
|
30
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Haug LS, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Jovanovic N, Philippat C, Eggesbo M, Lepage P, Slama R. Perinatal Exposure to Phenols and Poly- and Perfluoroalkyl Substances and Gut Microbiota in One-Year-Old Children. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15395-15414. [PMID: 39173114 DOI: 10.1021/acs.est.3c09927] [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: 08/24/2024]
Abstract
The role of the gut microbiota in human health calls for a better understanding of its determinants. In particular, the possible effects of chemicals with widespread exposure other than pharmaceuticals are little known. Our aim was to characterize the sensitivity of the early-life gut microbiota to specific chemicals with possible antimicrobial action. Within the SEPAGES French couple-child cohort, we assessed 12 phenols in repeated urine samples from 356 pregnant women and their offspring and 19 poly- and perfluoroalkyl substances (PFASs) in serum from the pregnant women. We collected stool samples from the children at one year of age, in which the V3-V4 region of the 16S rRNA gene was sequenced, allowing for gut bacterial profiling. Associations of each chemical with α- and β-diversity indices of the gut microbiota and with the relative abundance of the most abundant taxa were assessed using single-pollutant and mixture (BKMR) models. Perinatal exposure to certain parabens was associated with gut microbiota α- and β-diversity and with Firmicutes and Proteobacteria. Suggestive associations of certain phenols with genera of the Lachnospiraceae and Enterobacteriaceae families were observed, but these were not maintained after correction for multiple testing. Parabens, which have known antimicrobial properties, might disrupt the child gut microbiota, but larger studies are required to confirm these findings.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Line Småstuen Haug
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Nicolas Jovanovic
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Merete Eggesbo
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| |
Collapse
|
31
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
32
|
Jones K, Wessel LM, Schäfer KH, Tapia-Laliena MÁ. Use of Cosmetics in Pregnancy and Neurotoxicity: Can It Increase the Risk of Congenital Enteric Neuropathies? Biomolecules 2024; 14:984. [PMID: 39199372 PMCID: PMC11352589 DOI: 10.3390/biom14080984] [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: 07/02/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Pregnancy is a particularly vulnerable period for the growing fetus, when exposure to toxic agents, especially in the early phases, can decisively harm embryo development and compromise the future health of the newborn. The inclusion of various chemical substances in personal care products (PCPs) and cosmetic formulations can be associated with disruption and damage to the nervous system. Microplastics, benzophenones, parabens, phthalates and metals are among the most common chemical substances found in cosmetics that have been shown to induce neurotoxic mechanisms. Although cosmetic neurotoxin exposure is believed to be minimal, different exposure scenarios of cosmetics suggest that these neurotoxins remain a threat. Special attention should be paid to early exposure in the first weeks of gestation, when critical processes, like the migration and proliferation of the neural crest derived cells, start to form the ENS. Importantly, cosmetic neurotoxins can cross the placental barrier and affect the future embryo, but they are also secreted in breast milk, so babies remain exposed for longer periods, even after birth. In this review, we explore how neurotoxins contained in cosmetics and PCPs may have a role in the pathogenesis of various neurodevelopmental disorders and neurodegenerative diseases and, therefore, also in congenital enteric aganglionosis as well as in postnatal motility disorders. Understanding the mechanisms of these chemicals used in cosmetic formulations and their role in neurotoxicity is crucial to determining the safety of use for cosmetic products during pregnancy.
Collapse
Affiliation(s)
- Kendra Jones
- “Translational Medical Research” Master Program, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Lucas M. Wessel
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous Systems (AGENS), University of Applied Sciences Kaiserslautern, Amerikastrasse 1, 66482 Kaiserslautern, Germany;
| | - María Ángeles Tapia-Laliena
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
33
|
Bolat E, Sarıtaş S, Duman H, Eker F, Akdaşçi E, Karav S, Witkowska AM. Polyphenols: Secondary Metabolites with a Biological Impression. Nutrients 2024; 16:2550. [PMID: 39125431 PMCID: PMC11314462 DOI: 10.3390/nu16152550] [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: 07/05/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Polyphenols are natural compounds which are plant-based bioactive molecules, and have been the subject of growing interest in recent years. Characterized by multiple varieties, polyphenols are mostly found in fruits and vegetables. Currently, many diseases are waiting for a cure or a solution to reduce their symptoms. However, drug or other chemical strategies have limitations for using a treatment agent or still detection tool of many diseases, and thus researchers still need to investigate preventive or improving treatment. Therefore, it is of interest to elucidate polyphenols, their bioactivity effects, supplementation, and consumption. The disadvantage of polyphenols is that they have a limited bioavailability, although they have multiple beneficial outcomes with their bioactive roles. In this context, several different strategies have been developed to improve bioavailability, particularly liposomal and nanoparticles. As nutrition is one of the most important factors in improving health, the inclusion of plant-based molecules in the daily diet is significant and continues to be enthusiastically researched. Nutrition, which is important for individuals of all ages, is the key to the bioactivity of polyphenols.
Collapse
Affiliation(s)
- Ecem Bolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (E.B.); (S.S.); (H.D.); (F.E.); (E.A.)
| | - Anna Maria Witkowska
- Department of Food Biotechnology, Bialystok Medical University, 15-089 Bialystok, Poland
| |
Collapse
|
34
|
Ottria R, Xynomilakis O, Casati S, Ciuffreda P. Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases. Microorganisms 2024; 12:1582. [PMID: 39203424 PMCID: PMC11356122 DOI: 10.3390/microorganisms12081582] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Probiotics are "live microorganisms which, when administered in adequate amount, confer health benefits on the host". They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.
Collapse
Affiliation(s)
- Roberta Ottria
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, 20157 Milan, Italy; (O.X.); (S.C.); (P.C.)
| | | | | | | |
Collapse
|
35
|
Marik A, Biswas S, Banerjee ER. Exploring the relationship between gut microbial ecology and inflammatory disease: An insight into health and immune function. World J Immunol 2024; 14:96209. [DOI: 10.5411/wji.v14.i1.96209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024] Open
Abstract
The immune system, host brain development, and general metabolism are all influenced by the gut bacteria. Bacteria make up the majority of the gut microbiota in mammals. The mouse has been the most often used animal model in preclinical biological research. In mice, Firmicutes and Clostridiales are prominent. On the other hand, Bacteroidaceae, Prevotellaceae, and Firmicutes are commonly found in humans. In this review, we performed a detailed study by focusing on a comparison between human and murine gut microbiomes, role of the microbiome and their secreted metabolites in regulating gut immunity to maintain homeostasis, and changes in the microbial composition in the dysbiotic state.
Collapse
Affiliation(s)
- Akashlina Marik
- Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Saheli Biswas
- Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Ena Ray Banerjee
- Department of Zoology, University of Calcutta, Kolkata 700019, West Bengal, India
| |
Collapse
|
36
|
Wang J, Liu N, Chen Y, You J, Yang Y, Jin Y, Sun G, Zhou J. Knowledge, attitude, and practice of Chinese parents with infants (aged 0-3 years) toward immunity, gut microbiota and biotics: a comprehensive study. Front Immunol 2024; 15:1396087. [PMID: 39076968 PMCID: PMC11284022 DOI: 10.3389/fimmu.2024.1396087] [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: 03/05/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
In the wake of the COVID-19 pandemic, there has been an increasing focus towards infant immunity. The development and maintenance of the immune system are significantly influenced from birth, and is shaped by early-life infant feeding behavior. Hence, the knowledge, attitude, and practice (KAP) of parents play a crucial role in shaping the immune system of infants. A total of 2369 parents across 19 cities in China were surveyed using a self-designed online questionnaire. The KAP questionnaire assessed three domains: immunity, gut microbiota, and biotics (prebiotics, probiotics, and synbiotics). The questionnaire also included questions on infant health. An overwhelming majority of parents (97.90%) placed high value on their children's immunity, and 40.40% of them reported an increased level of concern following the COVID-19 pandemic. Diarrhea (78.80%), colds (75.70%), nighttime crying (73.80%), regurgitation (71.70%) and food retention (66.50%) were the major health issues reported. Knowledge scores toward immunity and gut microbiota were positively correlated to attitude and practice scores, respectively. Attitude scores toward immunity, gut microbiota and biotics were positively correlated to their respective practice scores. Parental knowledge score on immunity was negatively correlated with diarrhea, colds, regurgitation and food retention. On the other hand, parental practice scores toward immunity were negatively correlated with food retention. In regards to gut microbiota, parental knowledge score was negatively correlated with diarrhea, regurgitation and food retention; parental attitude score was negatively correlated with nighttime crying whereas practice score was negatively correlated with diarrhea, regurgitation, food retention and nighttime crying. Attitude score toward biotics was negatively correlated with nighttime crying and practice scores toward biotics was negatively correlated with colds, food retention and nighttime crying. This study demonstrated that significant gaps and misunderstandings exist among parents regarding immunity, gut microbiota health, and biotics. Both public education and interventions are crucial to enhance parental knowledge and practices, thereby improving infant immunity.
Collapse
Affiliation(s)
- Jiongnan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Nan Liu
- Chinese Nutrition Center for Education, Education and Training Center of Chinese Nutrition Society, Beijing, China
| | - Yufan Chen
- Danone Open Science Research Center for Life-Transforming Nutrition, Shanghai, China
| | - Jialu You
- Danone Open Science Research Center for Life-Transforming Nutrition, Shanghai, China
| | - Yunqing Yang
- Danone Open Science Research Center for Life-Transforming Nutrition, Shanghai, China
| | - Yi Jin
- Danone Open Science Research Center for Life-Transforming Nutrition, Shanghai, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Jin Zhou
- Chinese Nutrition Center for Education, Education and Training Center of Chinese Nutrition Society, Beijing, China
| |
Collapse
|
37
|
Hu Y, Zhang R, Li J, Wang H, Wang M, Ren Q, Fang Y, Tian L. Association Between Gut and Nasal Microbiota and Allergic Rhinitis: A Systematic Review. J Asthma Allergy 2024; 17:633-651. [PMID: 39006241 PMCID: PMC11246088 DOI: 10.2147/jaa.s472632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Allergic rhinitis is a chronic non-infectious inflammation of the nasal mucosa mediated by specific IgE. Recently, the human microbiome has drawn broad interest as a potential new target for treating this condition. This paper succinctly summarizes the main findings of 17 eligible studies published by February 2024, involving 1044 allergic rhinitis patients and 954 healthy controls from 5 countries. These studies examine differences in the human microbiome across important mucosal interfaces, including the nasal and intestinal areas, between patients and controls. Overall, findings suggest variations in the gut microbiota between allergic rhinitis patients and healthy individuals, although the specific bacterial taxa that significantly changed were not always consistent across studies. Due to the limited scope of existing research and patient coverage, the relationship between the nasal microbiome and allergic rhinitis remains inconclusive. The article discusses the potential immune-regulating role of the gut microbiome in allergic rhinitis. Further well-designed clinical trials with large-scale recruitment of allergic rhinitis patients are encouraged.
Collapse
Affiliation(s)
- Yucheng Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Rong Zhang
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Junjie Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Huan Wang
- Chengdu university of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Meiya Wang
- Chengdu university of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Qiuyi Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Yueqi Fang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Li Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
38
|
Ku S, Haque MA, Jang MJ, Ahn J, Choe D, Jeon JI, Park MS. The role of Bifidobacterium in longevity and the future of probiotics. Food Sci Biotechnol 2024; 33:2097-2110. [PMID: 39130652 PMCID: PMC11315853 DOI: 10.1007/s10068-024-01631-y] [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: 03/25/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 08/13/2024] Open
Abstract
This review explores the role and health impacts of probiotics, focusing specifically on Bifidobacterium spp. It highlights the functionalities that Bifidobacteria can provide, underscored by the historical evolution of definitions and technological advancements related to probiotics. By examining the association between Bifidobacteria and longevity, this review suggests new avenues for health enhancement. Highlighting case studies of centenarians, it presents examples related to human aging, illuminating the potential links to longevity through research on Bifidobacterium strains found in centenarians. This review not only emphasizes the importance of current research but also advocates for further investigation into the health benefits of Bifidobacteria, underlining the necessity for continuous study in the nutraceutical field.
Collapse
Affiliation(s)
- Seockmo Ku
- Department of Food Science and Technology, Texas A&M University, College Station, TX 77843 USA
| | - Md Ariful Haque
- Department of Food Science and Technology, Texas A&M University, College Station, TX 77843 USA
| | - Min Ji Jang
- Department of Food Science and Technology, Texas A&M University, College Station, TX 77843 USA
| | - Jaehyun Ahn
- Department of Agricultural Leadership, Education and Communications, Texas A&M University, College Station, TX 77843 USA
| | - Deokyeong Choe
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566 Korea
| | - Jong Ik Jeon
- Research Center, BIFIDO Co., Ltd, Hongcheon, 25117 South Korea
| | - Myeong Soo Park
- Research Center, BIFIDO Co., Ltd, Hongcheon, 25117 South Korea
| |
Collapse
|
39
|
Emokpae I, Tobia DL, Stamm SD, Lundy P, Weimer DS, Demory Beckler M. Examining the Efficacy of Five Lactobacillus Species in Treating and Preventing Atopic Dermatitis: A Systemic Literature Review. Cureus 2024; 16:e64833. [PMID: 39156317 PMCID: PMC11330270 DOI: 10.7759/cureus.64833] [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: 04/02/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Probiotics have garnered increasing attention, particularly within the realm of atopic dermatitis (AD). Although classified as dietary supplements by the Food and Drug Administration, probiotics are being explored for their potential to modify immune system responses and aid in disease recovery. This review aims to provide a current understanding of probiotics, specifically various lactobacilli strains, as a therapeutic option in preventing and treating AD. The concept of the gut-skin axis has gained substantial recognition, emphasizing the complex relationship between the gut microbiome and skin health. Dysfunctional gut barriers and metabolites produced by gut microorganisms can exert profound influences on skin conditions, including AD. Lactobacilli species are particularly noteworthy for their resilience and stability within the gastrointestinal tract, making these bacteria ideal candidates for probiotic supplementation. Various lactobacilli strains (Lactobacillus salivarius, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus reuteri, and Lactobacillus rhamnosus) were included in this study due to their current uses in mitigating AD symptomatology. This systemic review article aims to shed light on the potential of probiotics as a therapeutic approach for AD, highlighting their stellar safety profile and promising therapeutic efficacy. Given the compelling preliminary findings and the constraints associated with conventional treatments, probiotics, particularly lactobacilli strains, emerge as a considerable alternative or adjuvant option for individuals grappling with AD. Further exploration is imperative to establish probiotics as a promising therapeutic option, providing renewed hope for those seeking effective strategies for managing AD.
Collapse
Affiliation(s)
- Imina Emokpae
- Medical School, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Deanna L Tobia
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Saskia D Stamm
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Petra Lundy
- Family and Community Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Derek S Weimer
- Medical School, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
40
|
Fukuda R, Pak K, Kiuchi M, Hirata N, Mochimaru N, Tanaka R, Mitsui M, Ohya Y, Yoshida K. Longitudinal Correlations between Molecular Compositions of Stratum Corneum and Breast Milk Factors during Infancy: A Prospective Birth Cohort Study. Nutrients 2024; 16:1897. [PMID: 38931252 PMCID: PMC11206726 DOI: 10.3390/nu16121897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Breast milk contains numerous factors that are involved in the maturation of the immune system and development of the gut microbiota in infants. These factors include transforming growth factor-β1 and 2, immunoglobin A, and lactoferrin. Breast milk factors may also affect epidermal differentiation and the stratum corneum (SC) barrier in infants, but no studies examining these associations over time during infancy have been reported. In this single-center exploratory study, we measured the molecular components of the SC using confocal Raman spectroscopy at 0, 1, 2, 6, and 12 months of age in 39 infants born at our hospital. Breast milk factor concentrations from their mothers' breast milk were determined. Correlation coefficients for the two datasets were estimated for each molecular component of the SC and breast milk factor at each age and SC depth. The results showed that breast milk factors and molecular components of the SC during infancy were partly correlated with infant age in months and SC depth, suggesting that breast milk factors influence the maturation of the SC components. These findings may improve understanding of the pathogenesis of skin diseases associated with skin barrier abnormalities.
Collapse
Affiliation(s)
- Risa Fukuda
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Kyongsun Pak
- Division of Biostatistics, Department of Data Management, Center of Clinical Research and Development, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Megumi Kiuchi
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Hirata
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Mochimaru
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Ryo Tanaka
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Mari Mitsui
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kazue Yoshida
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
41
|
Tochitani S, Tsukahara T, Inoue R. Perturbed maternal microbiota shapes offspring microbiota during early colonization period in mice. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:335-352. [PMID: 38692912 PMCID: PMC11377213 DOI: 10.2183/pjab.100.020] [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: 05/03/2024]
Abstract
Recent studies have highlighted the impact of disrupted maternal gut microbiota on the colonization of offspring gut microbiota, with implications for offspring developmental trajectories. The extent to which offspring inherit the characteristics of altered maternal gut microbiota remains elusive. In this study, we employed a mouse model where maternal gut microbiota disruption was induced using non-absorbable antibiotics. Systematic chronological analyses of dam fecal samples, offspring luminal content, and offspring gut tissue samples revealed a notable congruence between offspring gut microbiota profiles and those of the perturbed maternal gut microbiota, highlighting the profound influence of maternal microbiota on early-life colonization of offspring gut microbiota. Nonetheless, certain dominant bacterial genera in maternal microbiota did not transfer to the offspring, indicating a bacterial taxonomy-dependent mechanism in the inheritance of maternal gut microbiota. Our results embody the vertical transmission dynamics of disrupted maternal gut microbiota in an animal model, where the gut microbiota of an offspring closely mirrors the gut microbiota of its mother.
Collapse
Affiliation(s)
- Shiro Tochitani
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
- Division of Development of Mental Functions, Research Center for Child Mental Development, University of Fukui, Fukui, Japan
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | | | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Hirakata, Osaka, Japan
| |
Collapse
|
42
|
Sun B, Sun Y, Sun Y, Zhou X, Han X, Han Y, Ma Q. Leucine Supplementation Modulates Lipid Metabolism and Inflammation in Early Weaning Piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38842880 DOI: 10.1021/acs.jafc.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Early weaning can induce the programmed dysregulation of glycolipid metabolism and inflammation in adult animals. The primary objective of this study was to evaluate the efficacy of leucine supplementation administered promptly after early weaning in mitigating these adverse effects in piglets. At day 21, 24 piglets were randomly selected and divided into 3 groups: EW group where the piglets were weaned at day 21 and fed basal diet, EWL group where the piglets were weaned at day 21 and fed the basal diet with supplementation of 1% leucine, and C group where the piglets were fed basal diet and weaned at 28 days. Each group contained eight replicates, with one piglet per replicate. The results indicated that early weaning had an impact on gut health and could activate the inhibitor of the kappa B kinase gamma/inhibitor kappa B alpha/NF-kappa-B (IKKγ/IκBα/NF-κB) signaling pathway to ameliorate pro-inflammatory factor and apoptosis levels. Furthermore, early weaning reduced the activity of fatty acid β oxidation (FAβO) and affected genes linked with lipid metabolism. Supplementing with leucine can improve the effects of these factors. In summary, leucine may alleviate the influences of early weaning on the lipid metabolism and inflammation in piglets.
Collapse
Affiliation(s)
- Bo Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yuchen Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yutong Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xinbo Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xuesong Han
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yixin Han
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
43
|
Kallio S, Jian C, Korpela K, Kukkonen AK, Salonen A, Savilahti E, Kuitunen M, M. de Vos W. Early-life gut microbiota associates with allergic rhinitis during 13-year follow-up in a Finnish probiotic intervention cohort. Microbiol Spectr 2024; 12:e0413523. [PMID: 38687061 PMCID: PMC11324021 DOI: 10.1128/spectrum.04135-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/06/2024] [Indexed: 05/02/2024] Open
Abstract
Perinatal and early-life factors reported to affect risk of allergic diseases may be mediated by changes in the gut microbiota. Here, we explored the associations between the infant gut microbiota and allergic morbidity in childhood until 13 years of age in a subgroup of the FLORA probiotic intervention cohort. A mixture of four probiotic strains with galacto-oligosaccharides was administrated to the mothers from the 36th week of the pregnancy and later to their infants until 6 months of age. The infants were monitored for the manifestations of atopic eczema, food allergy, allergic rhinitis, and asthma by a pediatrician at 2 and 5 years of age; the allergic status was subsequently verified by a questionnaire at 10 and 13 years of age. The fecal microbiota at 3 months was profiled by 16S rRNA amplicon sequencing targeting the V3-V4 region, with and without adjusting for potentially important early-life factors. Overall, the positive diagnosis for allergic rhinitis between 2 and 13 years was associated with microbiota composition both in non-adjusted and adjusted models. This association was more pronounced in children born to one parent with confirmed atopic diseases compared to those who had two atopic parents and was characterized by a lower relative abundance of Bifidobacterium and Escherichia/Shigella spp. and a higher proportion of Bacteroides. While the probiotic and galacto-oligosaccharides intervention in the entire cohort was previously shown to reduce the prevalence of eczema to a certain extent, no associations were found between the 3-month gut microbiota and childhood eczema in the studied sub-cohort.IMPORTANCEAllergic diseases have increased in prevalence during the past decades globally. Although probiotics have been considered a promising strategy for preventing certain allergy related symptoms, studies connecting the infant gut microbiota and later life allergic morbidity in various populations remain limited. The present study supports an association between the infant microbiota and allergic morbidity after first years of life, which has been rarely examined.CLINICAL TRIALSRegistered at ClinicalTrials.gov (NCT00298337).
Collapse
Affiliation(s)
- Sampo Kallio
- Children’s
Hospital, University of Helsinki and Helsinki University
Hospital, Helsinki,
Finland
| | - Ching Jian
- Human Microbiome
Research Program, Faculty of Medicine, University of
Helsinki, Helsinki,
Finland
| | - Katri Korpela
- Human Microbiome
Research Program, Faculty of Medicine, University of
Helsinki, Helsinki,
Finland
| | - Anna Kaarina Kukkonen
- Children’s
Hospital, University of Helsinki and Helsinki University
Hospital, Helsinki,
Finland
| | - Anne Salonen
- Human Microbiome
Research Program, Faculty of Medicine, University of
Helsinki, Helsinki,
Finland
| | - Erkki Savilahti
- Children’s
Hospital, University of Helsinki and Helsinki University
Hospital, Helsinki,
Finland
| | - Mikael Kuitunen
- Children’s
Hospital, University of Helsinki and Helsinki University
Hospital, Helsinki,
Finland
| | - Willem M. de Vos
- Human Microbiome
Research Program, Faculty of Medicine, University of
Helsinki, Helsinki,
Finland
- Laboratory of
Microbiology, Wageningen University,
Wageningen, the Netherlands
| |
Collapse
|
44
|
Newman NS, Abbott CA, Brame JE, Cando-Dumancela C, Fickling NW, Liddicoat C, Robinson JM, Breed MF. Childcare centre soil microbiomes are influenced by substrate type and surrounding vegetation condition. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172158. [PMID: 38583619 DOI: 10.1016/j.scitotenv.2024.172158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/25/2024] [Accepted: 03/31/2024] [Indexed: 04/09/2024]
Abstract
Urban development has profoundly reduced human exposure to biodiverse environments, which is linked to a rise in human disease. The 'biodiversity hypothesis' proposes that contact with diverse microbial communities (microbiota) benefits human health, as exposure to microbial diversity promotes immune training and regulates immune function. Soils and sandpits in urban childcare centres may provide exposure to diverse microbiota that support immunoregulation at a critical developmental stage in a child's life. However, the influence of outdoor substrate (i.e., sand vs. soil) and surrounding vegetation on these environmental microbiota in urban childcare centres remains poorly understood. Here, we used 16S rRNA amplicon sequencing to examine the variation in bacterial communities in sandpits and soils across 22 childcare centres in Adelaide, Australia, plus the impact of plant species richness and habitat condition on these bacterial communities. We show that sandpits had distinct bacterial communities and lower alpha diversity than soils. In addition, we found that plant species richness in the centres' yards and habitat condition surrounding the centres influenced the bacterial communities in soils but not sandpits. These results demonstrate that the diversity and composition of childcare centre sandpit and soil bacterial communities are shaped by substrate type, and that the soils are also shaped by the vegetation within and surrounding the centres. Accordingly, there is potential to modulate the exposure of children to health-associated bacterial communities by managing substrates and vegetation in and around childcare centres.
Collapse
Affiliation(s)
- Natalie S Newman
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Catherine A Abbott
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Joel E Brame
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | | | - Nicole W Fickling
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Craig Liddicoat
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Jake M Robinson
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Martin F Breed
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia.
| |
Collapse
|
45
|
Abraham JO, Lin B, Miller AE, Henry LP, Demmel MY, Warungu R, Mwangi M, Lobura PM, Pallares LF, Ayroles JF, Pringle RM, Rubenstein DI. Determinants of microbiome composition: Insights from free-ranging hybrid zebras (Equus quagga × grevyi). Mol Ecol 2024; 33:e17370. [PMID: 38682799 DOI: 10.1111/mec.17370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
The composition of mammalian gut microbiomes is highly conserved within species, yet the mechanisms by which microbiome composition is transmitted and maintained within lineages of wild animals remain unclear. Mutually compatible hypotheses exist, including that microbiome fidelity results from inherited dietary habits, shared environmental exposure, morphophysiological filtering and/or maternal effects. Interspecific hybrids are a promising system in which to interrogate the determinants of microbiome composition because hybrids can decouple traits and processes that are otherwise co-inherited in their parent species. We used a population of free-living hybrid zebras (Equus quagga × grevyi) in Kenya to evaluate the roles of these four mechanisms in regulating microbiome composition. We analysed faecal DNA for both the trnL-P6 and the 16S rRNA V4 region to characterize the diets and microbiomes of the hybrid zebra and of their parent species, plains zebra (E. quagga) and Grevy's zebra (E. grevyi). We found that both diet and microbiome composition clustered by species, and that hybrid diets and microbiomes were largely nested within those of the maternal species, plains zebra. Hybrid microbiomes were less variable than those of either parent species where they co-occurred. Diet and microbiome composition were strongly correlated, although the strength of this correlation varied between species. These patterns are most consistent with the maternal-effects hypothesis, somewhat consistent with the diet hypothesis, and largely inconsistent with the environmental-sourcing and morphophysiological-filtering hypotheses. Maternal transmittance likely operates in conjunction with inherited feeding habits to conserve microbiome composition within species.
Collapse
Affiliation(s)
- Joel O Abraham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Bing Lin
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- School of Public and International Affairs, Princeton University, Princeton, New Jersey, USA
| | - Audrey E Miller
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Lucas P Henry
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- Department of Biology, New York University, New York City, New York, USA
| | - Margaret Y Demmel
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- Section of Ecology, Behavior and Evolution, University of California San Diego, San Diego, California, USA
| | | | | | | | - Luisa F Pallares
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
- Friedrich Miescher Laboratory, Max Planck Society, Tübingen, Germany
| | - Julien F Ayroles
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Robert M Pringle
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - Daniel I Rubenstein
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
- Mpala Research Conservancy, Laikipia County, Kenya
| |
Collapse
|
46
|
Vasiliu O. Current evidence and future perspectives in the exploration of sleep-related eating disorder-a systematic literature review. Front Psychiatry 2024; 15:1393337. [PMID: 38873533 PMCID: PMC11169790 DOI: 10.3389/fpsyt.2024.1393337] [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: 02/28/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Sleep-related eating disorder (SRED) is a non-REM parasomnia with potentially significant negative effects on general health (dangerous activities during night eating episodes, obesity, or metabolic syndrome, for example). Although the history of SRED encompasses more than six decades, public awareness and even the awareness of the mental health specialists of this disorder is very limited, a phenomenon that hinders the development of research in this field. Therefore, a systematic review based on PRISMA 2020 guidelines explored the available evidence for SRED found in four electronic databases (PubMed, Cochrane Collaboration, Google Scholar, and Clarivate/Web of Science). A number of 94 primary and secondary reports were retrieved, investigating aspects regarding the risk factors, epidemiology, clinical data and differential diagnosis, epidemiology, structured evaluation, and treatment of SRED. Based on the results of these reports, Z-drugs, but also certain benzodiazepines, antidepressants, antipsychotics, and psychostimulants may trigger the onset of SRED. Psychiatric and neurologic disorders have also been associated with SRED, either as risk factors or comorbid conditions. Cerebral glucose metabolism dysfunctions, neurotransmitter dysfunctions, and genetic factors have been invoked as pathogenetic contributors. Structured assessment of SRED is possible, but there is a dearth of instruments dedicated to this purpose. Data on the prevalence and treatment of SRED exist, but good-quality epidemiological studies and clinical trials are still missing. In conclusion, future research is expected to address the shortcomings of SRED exploration by creating the conditions for better quality and larger group clinical research. The need for such investigation is granted by the importance of this pathology and its negative functional consequences.
Collapse
Affiliation(s)
- Octavian Vasiliu
- Department of Psychiatry, Dr. Carol Davila University Emergency Central Military Hospital, Bucharest, Romania
| |
Collapse
|
47
|
Kelly AM, McCarthy KN, Claxton TJ, Carlile SR, O'Brien EC, Vozza EG, Mills KH, McLoughlin RM. IL-10 inhibition during immunization improves vaccine-induced protection against Staphylococcus aureus infection. JCI Insight 2024; 9:e178216. [PMID: 38973612 PMCID: PMC11383370 DOI: 10.1172/jci.insight.178216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.
Collapse
Affiliation(s)
| | - Karen N McCarthy
- Host-Pathogen Interactions Group and
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Kingston Hg Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
48
|
Zhang M, Qiao H, Yang S, Kwok LY, Zhang H, Zhang W. Human Breast Milk: The Role of Its Microbiota and Metabolites in Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10665-10678. [PMID: 38691667 DOI: 10.1021/acs.jafc.3c07690] [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: 05/03/2024]
Abstract
This review explores the role of microorganisms and metabolites in human breast milk and their impact on neonatal health. Breast milk serves as both a primary source of nutrition for newborns and contributes to the development and maturation of the digestive, immunological, and neurological systems. It has the potential to reduce the risks of infections, allergies, and asthma. As our understanding of the properties of human milk advances, there is growing interest in incorporating its benefits into personalized infant nutrition strategies, particularly in situations in which breastfeeding is not an option. Future infant formula products are expected to emulate the composition and advantages of human milk, aligning with an evolving understanding of infant nutrition. The long-term health implications of human milk are still under investigation.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hui Qiao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shuwei Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
49
|
Hao D, Niu H, Zhao Q, Shi J, An C, Wang S, Zhou C, Chen S, Fu Y, Zhang Y, He Z. Impact of high-altitude acclimatization and de-acclimatization on the intestinal microbiota of rats in a natural high-altitude environment. Front Microbiol 2024; 15:1371247. [PMID: 38774503 PMCID: PMC11106481 DOI: 10.3389/fmicb.2024.1371247] [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: 01/19/2024] [Accepted: 04/18/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Intestinal microorganisms play an important role in the health of both humans and animals, with their composition being influenced by changes in the host's environment. Methods We evaluated the longitudinal changes in the fecal microbial community of rats at different altitudes across various time points. Rats were airlifted to high altitude (3,650 m) and acclimatized for 42 days (HAC), before being by airlifted back to low altitude (500 m) and de-acclimatized for 28 days (HADA); meanwhile, the control group included rats living at low altitude (500 m; LA). We investigated changes in the gut microbiota at 12 time points during high-altitude acclimatization and de-acclimatization, employing 16S rRNA gene sequencing technology alongside physiological indices, such as weight and daily autonomous activity time. Results A significant increase in the Chao1 index was observed on day 14 in the HAC and HADA groups compared to that in the LA group, indicating clear differences in species richness. Moreover, the principal coordinate analysis revealed that the bacterial community structures of HAC and HADA differed from those in LA. Long-term high-altitude acclimatization and de- acclimatization resulted in the reduced abundance of the probiotic Lactobacillus. Altitude and age significantly influenced intestinal microbiota composition, with changes in ambient oxygen content and atmospheric partial pressure being considered key causal factors of altitude-dependent alterations in microbiota composition. High-altitude may be linked to an increase in anaerobic bacterial abundance and a decrease in non-anaerobic bacterial abundance. Discussion In this study, the hypobaric hypoxic conditions at high-altitude increased the abundance of anaerobes, while reducing the abundance of probiotics; these changes in bacterial community structure may, ultimately, affect host health. Overall, gaining a comprehensive understanding of the intestinal microbiota alterations during high-altitude acclimatization and de-acclimatization is essential for the development of effective prevention and treatment strategies to better protect the health of individuals traveling between high- and low-altitude areas.
Collapse
Affiliation(s)
- Doudou Hao
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Haomeng Niu
- Medical College, Tibet University, Lhasa, China
| | - Qin Zhao
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Jing Shi
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Chuanhao An
- Health Clinic, Training Base of the Armed Police Force of Tibet, Lhasa, China
| | - Siyu Wang
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Chaohua Zhou
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Siyuan Chen
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Yongxing Fu
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Yongqun Zhang
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| | - Zeng He
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, China
| |
Collapse
|
50
|
Yu J, Zhang Y, Wells JCK, Wei Z, Bajaj-Elliott M, Nielsen DS, Fewtrell MS. A Stress Reduction Intervention for Lactating Mothers Alters Maternal Gut, Breast Milk, and Infant Gut Microbiomes: Data from a Randomized Controlled Trial. Nutrients 2024; 16:1074. [PMID: 38613107 PMCID: PMC11013067 DOI: 10.3390/nu16071074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND This secondary analysis of data from a randomized controlled trial (RCT) investigated how the maternal gut, breast milk, and infant gut microbiomes may contribute to the effects of a relaxation intervention, which reduced maternal stress and promoted infant weight gain. METHODS An RCT was undertaken in healthy Chinese primiparous mother-infant pairs (340/7-376/7gestation weeks). Mothers were randomly allocated to either the intervention group (IG, listening to relaxation meditation) or the control group (CG). Outcomes were the differences in microbiome composition and the diversity in the maternal gut, breast milk, and infant gut at 1 (baseline) and 8 weeks (post-intervention) between IG and CG, assessed using 16S rRNA gene amplicon sequencing of fecal and breastmilk samples. RESULTS In total, 38 mother-infant pairs were included in this analysis (IG = 19, CG = 19). The overall microbiome community structure in the maternal gut was significantly different between the IG and CG at 1 week, with the difference being more significant at 8 weeks (Bray-Curtis distance R2 = 0.04 vs. R2 = 0.13). Post-intervention, a significantly lower α-diversity was observed in IG breast milk (observed features: CG = 295 vs. IG = 255, p = 0.032); the Bifidobacterium genera presented a higher relative abundance. A significantly higher α-diversity was observed in IG infant gut (observed features: CG = 73 vs. IG = 113, p < 0.001). CONCLUSIONS The findings were consistent with the hypothesis that the microbiome might mediate observed relaxation intervention effects via gut-brain axis and entero-mammary pathways; but confirmation is required.
Collapse
Affiliation(s)
- Jinyue Yu
- Childhood Nutrition Research Group, Population, Policy & Practice Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.Y.); (J.C.K.W.)
| | - Yan Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Jonathan C. K. Wells
- Childhood Nutrition Research Group, Population, Policy & Practice Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.Y.); (J.C.K.W.)
| | - Zhuang Wei
- Department of Child Healthcare, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing 100045, China;
| | - Mona Bajaj-Elliott
- Infection, Immunity & Inflammation Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
| | | | - Mary S. Fewtrell
- Childhood Nutrition Research Group, Population, Policy & Practice Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.Y.); (J.C.K.W.)
| |
Collapse
|