1
|
Tüsüz Önata E, Özdemir Ö. Fecal microbiota transplantation in allergic diseases. World J Methodol 2025; 15:101430. [DOI: 10.5662/wjm.v15.i2.101430] [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: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024] Open
Abstract
Microorganisms such as bacteria, fungi, viruses, parasites living in the human intestine constitute the human intestinal microbiota. Dysbiosis refers to compositional and quantitative changes that negatively affect healthy gut microbiota. In recent years, with the demonstration that many diseases are associated with dysbiosis, treatment strategies targeting the correction of dysbiosis in the treatment of these diseases have begun to be investigated. Faecal microbiota transplantation (FMT) is the process of transferring faeces from a healthy donor to another recipient in order to restore the gut microbiota and provide a therapeutic benefit. FMT studies have gained popularity after probiotic, prebiotic, symbiotic studies in the treatment of dysbiosis and related diseases. FMT has emerged as a potential new therapy in the treatment of allergic diseases as it is associated with the maintenance of intestinal microbiota and immunological balance (T helper 1/T helper 2 cells) and thus suppression of allergic responses. In this article, the definition, application, safety and use of FMT in allergic diseases will be discussed with current data.
Collapse
Affiliation(s)
- Ece Tüsüz Önata
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| | - Öner Özdemir
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| |
Collapse
|
2
|
Wang L, Lai C, Yu J, Xu X, Jia M, Wang Z, Chen Y, Lou Q, Tao Q, Hu H, Fu Z, Jia X, Zhang W. Early-life antibiotic dysbiosis impairs microbial tryptophan- nicotinic acid metabolism exacerbating food allergy in adulthood. Int Immunopharmacol 2025; 159:114888. [PMID: 40403504 DOI: 10.1016/j.intimp.2025.114888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025]
Abstract
Food allergy (FA) pathogenesis links to intestinal dysbiosis, with antibiotic exposure a suspected risk factor, yet mechanisms are unclear. Our study shows early life (EL) antibiotic exposure in mice heightens susceptibility to OVA - induced allergic intestinal inflammation. EL - antibiotics cause intestinal dysbiosis, like Clostridia and Muribaculaceae depletion and Sutterellaceae enrichment, disrupting tryptophan metabolism and reducing nicotinic acid (NA). NA deficiency impairs gut barrier and Th2/Treg balance. However, NA supplementation restores these via GPR109A. In human pediatric cohorts, food - allergic children with EL - antibiotic exposure have lower gut NA levels. We integrated mouse and human data with multi - omics, revealing EL - Abx regulates FA through the "microbiota - metabolism - immunity" axis, and suggest targeting NA pathway to counter antibiotic - related FA risk.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Chuqiao Lai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Jiahui Yu
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Xinyi Xu
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Minghui Jia
- Zhejiang Chinese Medical University, Hangzhou City 310053, China
| | - Zheng Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Yeqing Chen
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Qianjin Lou
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Qihong Tao
- Wenzhou Medical University, Wenzhou City 325000, China
| | - Hao Hu
- Wenzhou Medical University, Wenzhou City 325000, China
| | - Zhanqing Fu
- Wenzhou Medical University, Wenzhou City 325000, China
| | - Xiaoxiao Jia
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou City 325000, China.
| |
Collapse
|
3
|
Liu X, Luo Y, Chen X, Wu M, Xu X, Tian J, Gao Y, Zhu J, Wang Z, Zhou Y, Zhang Y, Wang X, Li W, Lu Q, Yao X. Fecal microbiota transplantation against moderate-to-severe atopic dermatitis: A randomized, double-blind controlled explorer trial. Allergy 2025; 80:1377-1388. [PMID: 39470619 DOI: 10.1111/all.16372] [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: 04/05/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is a novel treatment for inflammatory diseases. Herein, we assess its safety, efficacy, and immunological impact in patients with moderate-to-severe atopic dermatitis (AD). METHODS In this randomized, double-blind, placebo-controlled clinical trial, we performed the efficacy and safety assessment of FMT for moderate-to-severe adult patients with AD. All patients received FMT or placebo once a week for 3 weeks, in addition to their standard background treatments. Patients underwent disease severity assessments at weeks 0, 1, 2, 4, 8, 12, and 16, and blood and fecal samples were collected for immunologic analysis and metagenomic shotgun sequencing, respectively. Safety was monitored throughout the trial. RESULTS Improvements in eczema area and severity index (EASI) scores and percentage of patients achieving EASI 50 (50% reduction in EASI score) were greater in patients treated with FMT than in placebo-treated patients. No serious adverse reactions occurred during the trial. FMT treatment decreased the Th2 and Th17 cell proportions among the peripheral blood mononuclear cells, and the levels of TNF-α, and total IgE in serum. By contrast, the expression levels of IL-12p70 and perforin on NK cells were increased. Moreover, FMT altered the abundance of species and functional pathways of the gut microbiota in the patients, especially the abundance of Megamonas funiformis and the pathway for 1,4-dihydroxy-6-naphthoate biosynthesis II. CONCLUSION FMT was a safe and effective therapy in moderate-to-severe adult patients with AD; the treatment changed the gut microbiota compositions and functions.
Collapse
Affiliation(s)
- Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yang Luo
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xingyu Chen
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Mingyang Wu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaoqiang Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jingru Tian
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yingxia Gao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jun Zhu
- 01life Institute, Shenzhen, China
| | | | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yu Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaokai Wang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Qianjin Lu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
4
|
Kelderer F, Granåsen G, Holmlund S, Silfverdal SA, Bamberg H, Mommers M, Penders J, Domellöf M, Mogren I, West CE. Respiratory morbidity before and during the COVID-19 pandemic from birth to 18 months in a Swedish birth cohort. J Allergy Clin Immunol 2025; 155:1214-1223.e10. [PMID: 39734033 DOI: 10.1016/j.jaci.2024.12.1080] [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: 04/22/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Respiratory infections in early life are an identified risk factor for asthma. We hypothesized that infection-prevention measures during the coronavirus disease 2019 (COVID-19) pandemic influenced the risk of respiratory morbidity and aeroallergen sensitization in early childhood. OBJECTIVE We compared respiratory morbidity and aeroallergen sensitization in children born before and during the pandemic. METHODS We compared a COVID-19 category (exposed children; n = 1661) to a pre-COVID-19 category (nonexposed children; n = 1676) by using data from the prospective population-based NorthPop Birth Cohort study in Sweden. Data on respiratory morbidity and concomitant medication were retrieved from national registers. Prospectively collected data on respiratory morbidity using web-based questionnaires at 9 and 18 months of age were applied. At age 18 months, serum IgE levels to aeroallergens were determined (n = 1702). RESULTS The risk of developing any respiratory tract infection (adjusted odds ratio [aOR] = 0.33 [95% CI, 0.26-0.42]), bronchitis (aOR = 0.50 [95% CI, 0.27-0.95]) and croup (aOR = 0.59 [95% CI, 0.37-0.94]) were decreased in the COVID-19 category. The risk of wheeze in the first 9 months was lower in the COVID-19 category (aOR = 0.70 [95% CI, 0.55-0.89]). There were also fewer prescriptions of antibiotics in the COVID-19 category. The prevalence of aeroallergen sensitization was similar between categories. CONCLUSION Children born during the COVID-19 pandemic demonstrated significantly decreased risks of respiratory infections and prescribed antibiotics until 18 months of age compared to children born before the COVID-19 pandemic. Whether this will affect the risk of developing asthma in childhood is being followed.
Collapse
Affiliation(s)
- Fanny Kelderer
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden.
| | - Gabriel Granåsen
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Sophia Holmlund
- Department of Nursing, Umeå University, Umeå, Sweden; Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden; Judith Lumley Centre, School of Nursing and Midwifery, La Trobe University, Bundoora, Australia
| | | | - Hilde Bamberg
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Monique Mommers
- Department of Epidemiology, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Ingrid Mogren
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Christina E West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| |
Collapse
|
5
|
Isoldi S, Mallardo S, Quitadamo P, Leter B, Cucchiara S. Review on Advances in Pediatric Endoscopy in the Management of Inflammatory Bowel Disease. Curr Pediatr Rev 2025; 21:154-165. [PMID: 38265388 DOI: 10.2174/0115733963268547231128101929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/26/2023] [Accepted: 10/19/2023] [Indexed: 01/25/2024]
Abstract
Over the past decades, an increased importance has been given to gastrointestinal (GI) endoscopy in the management of children with inflammatory bowel diseases (IBD), considering that mucosal healing has been recognized as the optimal endpoint in the treat-to-target paradigm. The recent advances in technology and anesthesia have facilitated the comprehensive evaluation of the GI tract. In this review, we will discuss the role of ileocolonoscopy, upper GI endoscopy, and device-assisted enteroscopy in the work-up and management of pediatric Crohn's disease (CD) and ulcerative colitis, with particular attention on non-invasive endoscopic techniques, such as wireless capsule endoscopy. We will also analyze the most commonly used endoscopic scoring systems, including small bowel scoring systems and endoscopic recurrence grading of neo-terminal ileum CD. Moreover, we will focus on the endoscopic management of complications, such as strictures, that commonly require surgery. Lastly, we will discuss cancer surveillance in children with IBD, with particular consideration of the role of high-definition endoscopic equipment and chromoendoscopy in dysplasia detection rates.
Collapse
Affiliation(s)
- Sara Isoldi
- Pediatric Gastroenterology and Hepatology Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
- Maternal and Child Health Department, Santa Maria Goretti Hospital, Sapienza-University of Rome, Latina, Italy
| | - Saverio Mallardo
- Maternal and Child Health Department, Santa Maria Goretti Hospital, Sapienza-University of Rome, Latina, Italy
| | - Paolo Quitadamo
- Pediatric Gastroenterology and Hepatology Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Beatrice Leter
- Department of Women's and Children's Health, Sapienza University of Rome, Rome, Italy
| | - Salvatore Cucchiara
- Department of Women's and Children's Health, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Gagliano C, Salvetat ML, Musa M, D'Esposito F, Rusciano D, Maniaci A, Pellegrini F, Scibilia G, Zeppieri M. Bacterial Insights: Unraveling the Ocular Microbiome in Glaucoma Pathogenesis. FRONT BIOSCI-LANDMRK 2024; 29:310. [PMID: 39206909 DOI: 10.31083/j.fbl2908310] [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: 04/11/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024]
Abstract
This review explores the connection between the ocular surface microbiome and glaucoma, highlighting its impact on disease progression. Beginning with an overview of global glaucoma significance, it emphasizes the importance of understanding the cellular characteristics and microbiology of the ocular microbiome. A search was conducted on the PubMed and Cochrane Library databases using the phrase "ocular microbiome glaucoma". 0 records were returned from the Cochrane Library while 21 were returned from PubMed. A total of 21 results were retrieved from 2017 to 2024. This comprised one opinion paper, four original research articles, and 16 reviews. This review covered the anatomy of the ocular surface, advanced analysis methods, and the ocular microbiome. It also delved into dysbiosis in glaucoma, addressing altered microbial communities and their potential role in disease progression. The intricate interplay between the ocular microbiome and the host's immune system is explored, emphasizing crosstalk and inflammatory responses. The review concludes by discussing therapeutic implications, including modulating ocular microbiota and potential future treatment strategies. Understanding the microbiome in healthy and glaucomatous eyes can help researchers and clinicians in innovative approaches to ocular health.
Collapse
Affiliation(s)
- Caterina Gagliano
- Department of Medicine and Surgery, University of Enna "Kore", 94100 Enna, Italy
- Eye Clinic, Catania University San Marco Hospital, 95121 Catania, Italy
| | - Maria Letizia Salvetat
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, 300238 Benin, Edo, Nigeria
| | - Fabiana D'Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, NW15QH London, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Napoli, Italy
| | | | - Antonino Maniaci
- Department of Medicine and Surgery, University of Enna "Kore", 94100 Enna, Italy
| | - Francesco Pellegrini
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Giuseppe Scibilia
- Department of Obstetrics and Gynecology, "Giovanni Paolo II" Hospital, 97100 Ragusa, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
7
|
Martemucci G, Khalil M, Di Luca A, Abdallah H, D’Alessandro AG. Comprehensive Strategies for Metabolic Syndrome: How Nutrition, Dietary Polyphenols, Physical Activity, and Lifestyle Modifications Address Diabesity, Cardiovascular Diseases, and Neurodegenerative Conditions. Metabolites 2024; 14:327. [PMID: 38921462 PMCID: PMC11206163 DOI: 10.3390/metabo14060327] [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: 04/21/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Several hallmarks of metabolic syndrome, such as dysregulation in the glucose and lipid metabolism, endothelial dysfunction, insulin resistance, low-to-medium systemic inflammation, and intestinal microbiota dysbiosis, represent a pathological bridge between metabolic syndrome and diabesity, cardiovascular, and neurodegenerative disorders. This review aims to highlight some therapeutic strategies against metabolic syndrome involving integrative approaches to improve lifestyle and daily diet. The beneficial effects of foods containing antioxidant polyphenols, intestinal microbiota control, and physical activity were also considered. We comprehensively examined a large body of published articles involving basic, animal, and human studie, as well as recent guidelines. As a result, dietary polyphenols from natural plant-based antioxidants and adherence to the Mediterranean diet, along with physical exercise, are promising complementary therapies to delay or prevent the onset of metabolic syndrome and counteract diabesity and cardiovascular diseases, as well as to protect against neurodegenerative disorders and cognitive decline. Modulation of the intestinal microbiota reduces the risks associated with MS, improves diabetes and cardiovascular diseases (CVD), and exerts neuroprotective action. Despite several studies, the estimation of dietary polyphenol intake is inconclusive and requires further evidence. Lifestyle interventions involving physical activity and reduced calorie intake can improve metabolic outcomes.
Collapse
Affiliation(s)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | - Alessio Di Luca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (A.D.L.); (A.G.D.)
| | - Hala Abdallah
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | | |
Collapse
|
8
|
Zheng X, Chen M, Zhuang Y, Xu J, Zhao L, Qian Y, Shen W. Genetic associations between gut microbiota and allergic rhinitis: an LDSC and MR analysis. Front Microbiol 2024; 15:1395340. [PMID: 38855765 PMCID: PMC11157438 DOI: 10.3389/fmicb.2024.1395340] [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: 03/03/2024] [Accepted: 04/26/2024] [Indexed: 06/11/2024] Open
Abstract
Background Several studies have suggested a potential link between allergic rhinitis (AR) and gut microbiota. In response, we conducted a meta-analysis of Linkage Disequilibrium Score Regression (LDSC) and Mendelian randomization (MR) to detect their genetic associations. Methods Summary statistics for 211 gut microbiota taxa were gathered from the MiBioGen study, while data for AR were sourced from the Pan-UKB, the FinnGen, and the Genetic Epidemiology Research on Aging (GERA). The genetic correlation between gut microbiota and AR was assessed using LDSC. The principal estimate of causality was determined using the Inverse-Variance Weighted (IVW) method. To assess the robustness of these findings, sensitivity analyses were conducted employing methods such as the weighted median, MR-Egger, and MR-PRESSO. The summary effect estimates of LDSC, forward MR and reverse MR were combined using meta-analysis for AR from different data resources. Results Our study indicated a significant genetic correlation between genus Sellimonas (Rg = -0.64, p = 3.64 × 10-5, Adjust_P = 3.64 × 10-5) and AR, and a suggestive genetic correlation between seven bacterial taxa and AR. Moreover, the forward MR analysis identified genus Gordonibacter, genus Coprococcus2, genus LachnospiraceaeUCG010, genus Methanobrevibacter, and family Victivallaceae as being suggestively associated with an increased risk of AR. The reverse MR analysis indicated that AR was suggestively linked to an increased risk for genus Coprococcus2 and genus RuminococcaceaeUCG011. Conclusion Our findings indicate a causal relationship between specific gut microbiomes and AR. This enhances our understanding of the gut microbiota's contribution to the pathophysiology of AR and lays the groundwork for innovative approaches and theoretical models for future prevention and treatment strategies in this patient population.
Collapse
Affiliation(s)
| | | | | | | | | | | | - WenMing Shen
- Emergency Department, Wujin People’s Hospital Affiliated with Jiangsu University and Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
9
|
Dang C, Chen Z, Chai Y, Liu P, Yu X, Liu Y, Liu J. Assessing the relationship between gut microbiota and endometriosis: a bidirectional two-sample mendelian randomization analysis. BMC Womens Health 2024; 24:123. [PMID: 38365715 PMCID: PMC10873948 DOI: 10.1186/s12905-024-02945-z] [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: 10/08/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND An increasing body of observational studies have indicated an association between gut microbiota and endometriosis. However, the causal relationship between them is not yet clear. In this study, we employed Mendelian randomization method to investigate the causal relationship between 211 gut microbiota taxa and endometriosis. METHODS Independent genetic loci significantly associated with the relative abundance of 211 gut microbiota taxa, based on predefined thresholds, were extracted as instrumental variables. The primary analytical approach employed was the IVW method. Effect estimates were assessed primarily using the odds ratio and 95% confidence intervals. Supplementary analyses were conducted using MR-Egger regression, the weighted median method, the simple mode and the weighted mode method to complement the IVW results. In addition, we conducted tests for heterogeneity, horizontal pleiotropy, sensitivity analysis, and MR Steiger to assess the robustness of the results and the strength of the causal relationships. RESULTS Based on the IVW method, we found that the family Prevotellaceae, genus Anaerotruncus, genus Olsenella, genus Oscillospira, and order Bacillales were identified as risk factors for endometriosis, while class Melainabacteria and genus Eubacterium ruminantium group were protective factors. Additionally, no causal relationship was observed between endometriosis and gut microbiota. Heterogeneity tests, pleiotropy tests, and leave-one-out sensitivity analyses did not detect any significant heterogeneity or pleiotropic effects. CONCLUSIONS Our MR study has provided evidence supporting a potential causal relationship between gut microbiota and endometriosis, and it suggests the absence of bidirectional causal effects. These findings could potentially offer new insights for the development of novel strategies for the prevention and treatment of endometriosis.
Collapse
Affiliation(s)
- Chunxiao Dang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Zhenting Chen
- Department of eugenic genetics, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, 257091, Shandong, China
| | - Yuyan Chai
- Department of obstetrics, The People's Hospital of Dongying Distric, Dongying, 257091, Shandong, China
| | - Pengfei Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Xiao Yu
- Department of gynaecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Yan Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
| | - Jinxing Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
10
|
Wang D, Russel WA, Macdonald KM, De Leon VM, Ay A, Belanger KD. Analysis of the gut microbiome in sled dogs reveals glucosamine- and activity-related effects on gut microbial composition. Front Vet Sci 2024; 11:1272711. [PMID: 38384960 PMCID: PMC10879321 DOI: 10.3389/fvets.2024.1272711] [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: 08/04/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
The composition of the microbiome influences many aspects of physiology and health, and can be altered by environmental factors, including diet and activity. Glucosamine is a dietary supplement often administered to address arthritic symptoms in humans, dogs, and other mammals. To investigate how gut microbial composition varies with glucosamine supplementation, we performed 16S rRNA sequence analysis of fecal samples from 24 Alaskan and Inuit huskies and used mixed effects models to investigate associations with activity, age, and additional factors. Glucosamine ingestion, age, activity, sex, and diet were correlated with differences in alpha-diversity, with diversity decreasing in dogs consuming glucosamine. Beta-diversity analysis revealed clustering of dogs based on glucosamine supplementation status. Glucosamine supplementation and exercise-related activity were associated with greater inter-individual pairwise distances. At the family level, Lactobacillaceae and Anaerovoracaceae relative abundances were lower in supplemented dogs when activity was accounted for. At the genus level, Eubacterium [brachy], Sellimonus, Parvibacter, and an unclassified genus belonging to the same family as Parvibacter (Eggerthellaceae) all were lower in supplemented dogs, but only significantly so post-activity. Our findings suggest that glucosamine supplementation alters microbiome composition in sled dogs, particularly in the context of exercise-related activity.
Collapse
Affiliation(s)
- Dong Wang
- Department of Computer Science, Colgate University, Hamilton, NY, United States
- Department of Mathematics, Colgate University, Hamilton, NY, United States
| | - William A. Russel
- Department of Biology, Colgate University, Hamilton, NY, United States
| | | | | | - Ahmet Ay
- Department of Mathematics, Colgate University, Hamilton, NY, United States
- Department of Biology, Colgate University, Hamilton, NY, United States
| | | |
Collapse
|
11
|
Monga N, Sharma S, Bhatia R, Bishnoi M, Kiran Kondepudi K, Naura AS. Immunomodulatory action of synbiotic comprising of newly isolated lactic acid producing bacterial strains against allergic asthma in mice. Cell Immunol 2023; 393-394:104786. [PMID: 37984277 DOI: 10.1016/j.cellimm.2023.104786] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/04/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Given the reported role of gut-microbiota in asthma pathogenesis, the present work was carried to evaluate immunomodulatory action of newly isolated lactic acid producing bacterial strains Bifidobacterium breve Bif11 and Lactiplantibacillus plantarum LAB31 against asthma using ovalbumin (OVA) based mouse model. Our results show that both strains modulate Th2 immune response potentially through production of short chain fatty acids (SCFAs), resulting in suppression of OVA-induced airway inflammation. Furthermore, synbiotic comprising of both strains and prebiotic, Isomaltooligosaccharide exhibited superior potential in amelioration of OVA-induced airway inflammation through improved modulation of Th2 immune response. Further, synbiotic protects against OVA-induced mucus hyper-production and airway-hyperresponsiveness. Such protection was associated with normalization of gut microbiome and enhanced production of SCFAs in cecum which correlates closely with population of T-regulatory cells in spleen. Overall, our novel synbiotic possesses the ability to fine-tune the immune response for providing protection against allergic asthma.
Collapse
Affiliation(s)
- Naina Monga
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Shikha Sharma
- Healthy Gut Research Group, Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, India; Adjunct Faculty, Department of Biotechnology, Panjab University, Chandigarh 160014, India
| | - Ruchika Bhatia
- Healthy Gut Research Group, Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, India; Adjunct Faculty, Department of Biotechnology, Panjab University, Chandigarh 160014, India
| | - Mahendra Bishnoi
- Healthy Gut Research Group, Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, India; Adjunct Faculty, Department of Biotechnology, Panjab University, Chandigarh 160014, India; Adjunct Faculty, Regional Centre for Biotechnology, Faridabad, India
| | - Kanthi Kiran Kondepudi
- Healthy Gut Research Group, Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, Mohali, India; Adjunct Faculty, Department of Biotechnology, Panjab University, Chandigarh 160014, India; Adjunct Faculty, Regional Centre for Biotechnology, Faridabad, India.
| | - Amarjit S Naura
- Department of Biochemistry, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
12
|
Fredriksson E, Bodén S, Domellöf M, West CE. Fruit Pouch Consumption Does Not Associate with Early Manifestations of Allergic Disease. Nutrients 2023; 15:4318. [PMID: 37892394 PMCID: PMC10609626 DOI: 10.3390/nu15204318] [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: 09/18/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Consumption of acidic fruit pouches in infancy may damage the epithelial barrier in the gastrointestinal tract and is suggested to increase allergy risk. We aimed to explore if a high fruit pouch consumption is associated with a higher incidence of early allergic manifestations. We included 2959 parent-child dyads from the Swedish prospective, population-based NorthPop birth cohort study with parentally reported data on frequency of fruit pouch consumption at 9 months of age, as well as parentally reported eczema, wheeze, physician-diagnosed asthma, and food allergy in the first 18 months of life. Immunoglobulin E levels (IgE) in serum (n = 1792), as response to a food mix and an inhalant mix, were determined at age 18 months. Compared with no consumption, daily consumption of one or more pouches at 9 months of age was associated with inhalant sensitization (odds ratio (OR) 2.27, 95% confidence interval (CI) 1.06-4.87, n = 1792) but did not remain significant in the multivariable adjusted model (aOR 2.08, 95% CI 0.95-4.53, n = 1679). There were no associations between fruit pouch consumption and allergic manifestations at this young age. This study suggests that fruit pouch consumption is not associated with allergic phenotypes or IgE sensitization in early childhood.
Collapse
Affiliation(s)
| | | | | | - Christina E. West
- Department of Clinical Sciences, Pediatrics, Umeå University, SE 90185 Umeå, Sweden; (E.F.); (S.B.); (M.D.)
| |
Collapse
|
13
|
Aziz C, Morales A, Pinto W, Fanchini V, Dell Aquila L, Sangaleti C, Elias R, Dalboni M. Evaluation of IL-6, FoxP3 Treg lymphocytes, intestinal barrier biomarkers and the use of synbiotics in obese adolescents: a pilot study. Front Pediatr 2023; 11:1215793. [PMID: 37859769 PMCID: PMC10583575 DOI: 10.3389/fped.2023.1215793] [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: 05/02/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
Aim This prospective pilot study evaluated inflammatory and intestinal barrier biomarkers and the effects of a synbiotic in obese adolescents. Methods Eighteen obese and 20 eutrophic adolescents were evaluated for body composition using bioimpedance analysis (BIA), body mass index (BMI), IL-6 and lipopolysaccharide (LPS) serum levels, CD4 and FoxP3 Treg lymphocytes and monocytes. Synbiotic supplementation for 60 days was also evaluated for these parameters only in obese adolescents. Results We observed an increase in CD4 lymphocyte (18.0 ± 12.4 vs. 8.9 ± 7.5; p < 0.01), IL-6 (0.30 ± 0.06 vs. 0.20 ± 0.06; p = 0.02) and LPS (0.18 ± 0.15 vs. 0.08 ± 0.05; p < 0.01) levels in obese compared to eutrophic adolescents. After synbiotic supplementation, FoxP3 Treg lymphocytes increased (14.0 ± 6.7 vs. 9.9 ± 5.4; p = 0.02) in obese adolescents. Conclusions Obese adolescents presented a state of microinflammation and intestinal barrier breakdown, and synbiotic supplementation increased the expression of FoxP3 Treg lymphocytes, an anti-inflammatory regulator. Whether the increase in FoxP3 Treg lymphocytes may have an impact on inflammation and outcomes in obese adolescents deserves further evaluation.
Collapse
Affiliation(s)
- Cylmara Aziz
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Armando Morales
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Walter Pinto
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Vanessa Fanchini
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Luis Dell Aquila
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Carine Sangaleti
- Department of Postgraduate Studies in Nanosciences and Biosciences, Universidade Estadual do Centro Oeste, Guarapuava, Brazil
| | - Rosilene Elias
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| | - Maria Dalboni
- Department of Postgraduate Studies in Medicine, Universidade Nove de Julho, São Paulo, Brazil
| |
Collapse
|
14
|
Chang C, Wang Q, Li X, Tan H, Huang G. The relationship between prebiotic intake and allergic rhinitis. Laryngoscope Investig Otolaryngol 2023; 8:1146-1153. [PMID: 37899852 PMCID: PMC10601581 DOI: 10.1002/lio2.1158] [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: 07/06/2023] [Revised: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Objectives Exploring the relationship between intake of probiotics and the prevalence of allergic rhinitis. Methods Based on data from the National Health and Nutrition Examination Survey, dietary supplement labels were examined to identify products containing probiotics and prebiotics. Statistical methods were used to analyze the factors influencing the prevalence of allergic rhinitis, and further stratified analysis was conducted to control for confounding factors. Results The proportion of individuals not consuming probiotics was significantly higher in the allergic rhinitis (AR) group than in those consuming them, suggesting a correlation between probiotics and AR. In the male subgroup with probiotic intake, the adjusted odds ratio (95% confidence interval) was 0.28 (0.10-0.75), p = .02, indicating that probiotic intake was a protective factor for AR in the male population. In the probiotic-intake group, the odds ratio for age < 65 was 0.26 (0.07-0.94), p = .04, and for age ≥ 80 was less than 1 with p < .0001, suggesting that probiotic intake was a protective factor for AR in age < 65 and age ≥ 80 populations, both with statistical significance. Conclusion Intake of probiotics is associated with a reduced prevalence of allergic rhinitis, particularly in the male population and individuals aged <65 years and ≥ 80 years. Level of Evidence Level 4.
Collapse
Affiliation(s)
- Chao Chang
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Qiuyang Wang
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Xiaodan Li
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Huazhang Tan
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Guoxin Huang
- Department of Evidence‐Based Medicine Center, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangChina
- School of Public Health and ManagementHubei University of MedicineShiyanHubeiChina
| |
Collapse
|
15
|
Spatz M, Wang Y, Lapiere A, Da Costa G, Michaudel C, Danne C, Michel ML, Langella P, Sokol H, Richard ML. Saccharomyces boulardii CNCM I-745 supplementation during and after antibiotic treatment positively influences the bacterial gut microbiota. Front Med (Lausanne) 2023; 10:1087715. [PMID: 37601783 PMCID: PMC10436532 DOI: 10.3389/fmed.2023.1087715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Antibiotic effects on gut bacteria have been widely studied, but very little is known about the consequences of such treatments on the mycobiota, the fungal part of the microbiota and how the length of administration influences both microbiota. Here, we examined the effect of antibiotics (ATB) on the composition of bacterial and fungal microbiota and how the administration of Saccharomyces boulardii CNCM I-745 influences both microbiota. Methods In order to get closer to the human microbiota, the mice used in this study were subjected to fecal microbiota transfer (FMT) using human feces and subsequently called human microbiotaassociated (HMA) mice. These mice were then treated with amoxicillinclavulanate antibiotics and supplemented with S. boulardii during and after ATB treatment to understand the effect of the yeast probiotic on both bacterial and fungal microbiota. Bacterial and fungal microbiota analyses were done using 16S and ITS2 rRNA amplicon-based sequencing. Results We showed that the administration of S. boulardii during ATB treatment had very limited effect on the fungal populations on the long term, once the yeast probiotic has been cleared from the gut. Concerning bacterial microbiota, S. boulardii administration allowed a better recovery of bacterial populations after the end of the ATB treatment period. Additionally, 16S and ITS2 rRNA sequence analysis revealed that 7 additional days of S. boulardii administration (17 days in total) enhanced the return of the initial bacterial equilibrium. Discussion In this study, we provide a comprehensive analysis of how probiotic yeast administration can influence the fungal and bacterial microbiota in a model of broad-spectrum antibiotherapy.
Collapse
Affiliation(s)
- Madeleine Spatz
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Yazhou Wang
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Alexia Lapiere
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Gregory Da Costa
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Chloé Michaudel
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Camille Danne
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Sorbonne Université, Paris, France
| | - Marie-Laure Michel
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Philippe Langella
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Harry Sokol
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Sorbonne Université, Paris, France
| | - Mathias L. Richard
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| |
Collapse
|
16
|
Srivastava K, Cao M, Fidan O, Shi Y, Yang N, Nowak-Wegrzyn A, Miao M, Zhan J, Sampson HA, Li XM. Berberine-containing natural-medicine with boiled peanut-OIT induces sustained peanut-tolerance associated with distinct microbiota signature. Front Immunol 2023; 14:1174907. [PMID: 37575233 PMCID: PMC10415201 DOI: 10.3389/fimmu.2023.1174907] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023] Open
Abstract
Background Gut microbiota influence food allergy. We showed that the natural compound berberine reduces IgE and others reported that BBR alters gut microbiota implying a potential role for microbiota changes in BBR function. Objective We sought to evaluate an oral Berberine-containing natural medicine with a boiled peanut oral immunotherapy (BNP) regimen as a treatment for food allergy using a murine model and to explore the correlation of treatment-induced changes in gut microbiota with therapeutic outcomes. Methods Peanut-allergic (PA) mice, orally sensitized with roasted peanut and cholera toxin, received oral BNP or control treatments. PA mice received periodic post-therapy roasted peanut exposures. Anaphylaxis was assessed by visualization of symptoms and measurement of body temperature. Histamine and serum peanut-specific IgE levels were measured by ELISA. Splenic IgE+B cells were assessed by flow cytometry. Fecal pellets were used for sequencing of bacterial 16S rDNA by Illumina MiSeq. Sequencing data were analyzed using built-in analysis platforms. Results BNP treatment regimen induced long-term tolerance to peanut accompanied by profound and sustained reduction of IgE, symptom scores, plasma histamine, body temperature, and number of IgE+ B cells (p <0.001 vs Sham for all). Significant differences were observed for Firmicutes/Bacteroidetes ratio across treatment groups. Bacterial genera positively correlated with post-challenge histamine and PN-IgE included Lachnospiraceae, Ruminococcaceae, and Hydrogenanaerobacterium (all Firmicutes) while Verrucromicrobiacea. Caproiciproducens, Enterobacteriaceae, and Bacteroidales were negatively correlated. Conclusions BNP is a promising regimen for food allergy treatment and its benefits in a murine model are associated with a distinct microbiota signature.
Collapse
Affiliation(s)
- Kamal Srivastava
- General Nutraceutical Technology, Elmsford, NY, United States
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Mingzhuo Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ozkan Fidan
- Department of Biological Engineering, Utah State University, Logan, UT, United States
- Department of Bioengineering, Abdullah Gul University, Kayseri, Türkiye
| | - Yanmei Shi
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Nan Yang
- General Nutraceutical Technology, Elmsford, NY, United States
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Anna Nowak-Wegrzyn
- Hassenfeld Children’s Hospital, Department of Pediatrics, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jixun Zhan
- Department of Biological Engineering, Utah State University, Logan, UT, United States
| | - Hugh A. Sampson
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
- Department of Otolaryngology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
17
|
Arzola-Martínez L, Ptaschinski C, Lukacs NW. Trained innate immunity, epigenetics, and food allergy. FRONTIERS IN ALLERGY 2023; 4:1105588. [PMID: 37304168 PMCID: PMC10251748 DOI: 10.3389/falgy.2023.1105588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
In recent years the increased incidence of food allergy in Western culture has been associated with environmental factors and an inappropriate immune phenotype. While the adaptive immune changes in food allergy development and progression have been well-characterized, an increase in innate cell frequency and activation status has also recently received greater attention. Early in prenatal and neonatal development of human immunity there is a reliance on epigenetic and metabolic changes that stem from environmental factors, which are critical in training the immune outcomes. In the present review, we discuss how trained immunity is regulated by epigenetic, microbial and metabolic factors, and how these factors and their impact on innate immunity have been linked to the development of food allergy. We further summarize current efforts to use probiotics as a potential therapeutic approach to reverse the epigenetic and metabolic signatures and prevent the development of severe anaphylactic food allergy, as well as the potential use of trained immunity as a diagnostic and management strategy. Finally, trained immunity is presented as one of the mechanisms of action of allergen-specific immunotherapy to promote tolerogenic responses in allergic individuals.
Collapse
Affiliation(s)
- Llilian Arzola-Martínez
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center (MHWFAC), University of Michigan, Ann Arbor, MI, United States
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center (MHWFAC), University of Michigan, Ann Arbor, MI, United States
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center (MHWFAC), University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
18
|
Freguia CF, Pascual DW, Fanger GR. Sjögren's Syndrome Treatments in the Microbiome Era. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230004. [PMID: 37323129 PMCID: PMC10270702 DOI: 10.20900/agmr20230004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by inflammatory cell infiltration of the salivary and lacrimal glands, resulting in acinar epithelial cell atrophy, cell death, and loss of exocrine function. At least half of SS patients develop extraglandular inflammatory disease and have a wide range of systemic clinical manifestations that can affect any organ system, including connective tissues. As many as 3.1 million people in the U.S. suffer from SS, a disease that causes severe impairment. Women are nine times more likely than men to be affected by this condition. Unfortunately, there is currently no effective treatment for SS, and the available options only provide partial relief. Treatment involves using replacement therapies such as artificial saliva and eye lubricants, or immunosuppressive agents that have limited efficacy. The medical community recognizes that there is a significant need for more effective treatments for SS. Increasing evidence demonstrates the links between the dysfunction of the human microbial community and the onset and development of many human diseases, signifying the potential use of microorganisms as an alternative strategy to conquer these issues. The role of the microbiome in controlling immune function of the human host in the context of autoimmune diseases like SS is now becoming better understood and may help to enable new drug development strategies. Natural probiotics and synthetic biology applications hold promise for novel treatment approaches to solve the encryption of many complex and multifactorial immune disorders, like SS.
Collapse
Affiliation(s)
| | - David W. Pascual
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611, USA
| | - Gary R. Fanger
- Rise Therapeutics, 1405 Research Blvd., Rockville, MD 20850, USA
| |
Collapse
|
19
|
Spatz M, Da Costa G, Ventin-Holmberg R, Planchais J, Michaudel C, Wang Y, Danne C, Lapiere A, Michel ML, Kolho KL, Langella P, Sokol H, Richard ML. Antibiotic treatment using amoxicillin-clavulanic acid impairs gut mycobiota development through modification of the bacterial ecosystem. MICROBIOME 2023; 11:73. [PMID: 37032359 PMCID: PMC10084673 DOI: 10.1186/s40168-023-01516-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 03/09/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Effects of antibiotics on gut bacteria have been widely studied, but very little is known about the consequences of such treatments on the fungal microbiota (mycobiota). It is commonly believed that fungal load increases in the gastrointestinal tract following antibiotic treatment, but better characterization is clearly needed of how antibiotics directly or indirectly affect the mycobiota and thus the entire microbiota. DESIGN We used samples from humans (infant cohort) and mice (conventional and human microbiota-associated mice) to study the consequences of antibiotic treatment (amoxicillin-clavulanic acid) on the intestinal microbiota. Bacterial and fungal communities were subjected to qPCR or 16S and ITS2 amplicon-based sequencing for microbiota analysis. In vitro assays further characterized bacterial-fungal interactions, with mixed cultures between specific bacteria and fungi. RESULTS Amoxicillin-clavulanic acid treatment triggered a decrease in the total fungal population in mouse feces, while other antibiotics had opposite effects on the fungal load. This decrease is accompanied by a total remodelling of the fungal population with the enrichment in Aspergillus, Cladosporium, and Valsa genera. In the presence of amoxicillin-clavulanic acid, microbiota analysis showed a remodeling of bacterial microbiota with an increase in specific bacteria belonging to the Enterobacteriaceae. Using in vitro assays, we isolated different Enterobacteriaceae species and explored their effect on different fungal strains. We showed that Enterobacter hormaechei was able to reduce the fungal population in vitro and in vivo through yet unknown mechanisms. CONCLUSIONS Bacteria and fungi have strong interactions within the microbiota; hence, the perturbation initiated by an antibiotic treatment targeting the bacterial community can have complex consequences and can induce opposite alterations of the mycobiota. Interestingly, amoxicillin-clavulanic acid treatment has a deleterious effect on the fungal community, which may have been partially due to the overgrowth of specific bacterial strains with inhibiting or competing effects on fungi. This study provides new insights into the interactions between fungi and bacteria of the intestinal microbiota and might offer new strategies to modulate gut microbiota equilibrium. Video Abstract.
Collapse
Affiliation(s)
- Madeleine Spatz
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France.
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France.
| | - Gregory Da Costa
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Rebecka Ventin-Holmberg
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, 00014, Helsinki, Finland
- Folkhälsan Research Center, 00250, Helsinki, Finland
| | - Julien Planchais
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Chloé Michaudel
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Yazhou Wang
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Camille Danne
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Alexia Lapiere
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Marie-Laure Michel
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Kaija-Leena Kolho
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, 00014, Helsinki, Finland
- Children's Hospital, Helsinki University, 00029, Helsinki, Finland
- Department of Pediatrics, Tampere University, 33520, Tampere, Finland
| | - Philippe Langella
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
| | - Harry Sokol
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France
- Gastroenterology Department, Centre de Recherche Saint-Antoine (CRSA), Saint Antoine Hospital, INSERM, Sorbonne Université, AP-HP, Paris, 75012, France
| | - Mathias L Richard
- Micalis Institute, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, AgroParisTech, France.
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, 75012, France.
| |
Collapse
|
20
|
Veeraraghavan B, Kesavelu D, Yadav B. Gut Microbiota Composition in Indian and Western Infants (0–24 Months): A Systematic Review. NUTRITION AND DIETARY SUPPLEMENTS 2023. [DOI: 10.2147/nds.s402256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
|
21
|
Sjödin KS, Sjödin A, Ruszczyński M, Kristensen MB, Hernell O, Szajewska H, West CE. Targeting the gut-lung axis by synbiotic feeding to infants in a randomized controlled trial. BMC Biol 2023; 21:38. [PMID: 36803508 PMCID: PMC9940374 DOI: 10.1186/s12915-023-01531-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/27/2023] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Formula-fed infants are at increased risk of infections. Due to the cross-talk between the mucosal systems of the gastrointestinal and respiratory tracts, adding synbiotics (prebiotics and probiotics) to infant formula may prevent infections even at distant sites. Infants that were born full term and weaned from breast milk were randomized to prebiotic formula (fructo- and galactooligosaccharides) or the same prebiotic formula with Lactobacillus paracasei ssp. paracasei F19 (synbiotics) from 1 to 6 months of age. The objective was to examine the synbiotic effects on gut microbiota development. RESULTS Fecal samples collected at ages 1, 4, 6, and 12 months were analyzed using 16S rRNA gene sequencing and a combination of untargeted gas chromatography-mass spectrometry/liquid chromatography-mass spectrometry. These analyses revealed that the synbiotic group had a lower abundance of Klebsiella, a higher abundance of Bifidobacterium breve compared to the prebiotic group, and increases in the anti-microbial metabolite d-3-phenyllactic acid. We also analyzed the fecal metagenome and antibiotic resistome in the 11 infants that had been diagnosed with lower respiratory tract infection (cases) and 11 matched controls using deep metagenomic sequencing. Cases with lower respiratory tract infection had a higher abundance of Klebsiella species and antimicrobial resistance genes related to Klebsiella pneumoniae, compared to controls. The results obtained using 16S rRNA gene amplicon and metagenomic sequencing were confirmed in silico by successful recovery of the metagenome-assembled genomes of the bacteria of interest. CONCLUSIONS This study demonstrates the additional benefit of feeding specific synbiotics to formula-fed infants over prebiotics only. Synbiotic feeding led to the underrepresentation of Klebsiella, enrichment of bifidobacteria, and increases in microbial degradation metabolites implicated in immune signaling and in the gut-lung and gut-skin axes. Our findings support future clinical evaluation of synbiotic formula in the prevention of infections and associated antibiotic treatment as a primary outcome when breastfeeding is not feasible. TRIAL REGISTRATION ClinicalTrials.gov NCT01625273 . Retrospectively registered on 21 June 2012.
Collapse
Affiliation(s)
- Kotryna Simonyté Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Andreas Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden ,Division of CBRN Security and Defense, FOI – Swedish Defense Research Agency, Umeå, Sweden
| | - Marek Ruszczyński
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | | - Olle Hernell
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Hania Szajewska
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Christina E. West
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| |
Collapse
|
22
|
Lif Holgerson P, Esberg A, West CE, Johansson I. The breast milk and childhood gastrointestinal microbiotas and disease outcomes: a longitudinal study. Pediatr Res 2023; 93:570-578. [PMID: 36216869 PMCID: PMC9988688 DOI: 10.1038/s41390-022-02328-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND We aimed to characterize breast milk microbiota and define associations with saliva and fecal microbiota and selected diseases in preschool children. METHODS In a longitudinal cohort study, the microbiotas from breast milk, mouth, and fecal samples were characterized by 16S rRNA gene sequencing. Questionnaires and medical records provided information on demographics, medical, and dental data. RESULTS The phylogeny in breast milk, saliva swabs, and feces differed at all levels (p < 0.0003), though all harbored species in Streptococcus, Veillonella, and Haemophilus. Species richness was highest in breast milk with increasing resemblance with the oral swab microbiota by increasing age. Caries-affected children at age 5 had been fed breast milk with tenfold higher abundance of caries-associated bacteria, e.g., Streptococcus mutans, than caries-free children (p < 0.002). At that age, taxa, e.g., Neisseria sicca were overrepresented in saliva swabs of children with otitis media (LDA score >2, p < 0.05). Gut symbionts, e.g., Bacteroides, were underrepresented in 3-month fecal samples in children later diagnosed with allergic disease (LDA score >2, p < 0.05). CONCLUSIONS Distinct microbiotas for the three sources were confirmed, though resemblance between milk and oral swab microbiota increased by age. Future studies should evaluate if the observed associations with disease outcomes are causal. IMPACT Few studies have studied the association between breast milk microbiota and gastrointestinal microbiota beyond early infancy. The present study confirms distinct microbiota profiles in breast milk, saliva swabs, and feces in infancy and indicates increasing resemblance between breast milk and the oral microbiota by increasing age. The fecal microbiota at 3 months was associated with later allergic disease; the saliva microbiota by age 5 differed between children with and without otitis media at the same age; and children with caries by age 5 had been fed breast milk with a higher abundance of caries-associated bacteria.
Collapse
Affiliation(s)
- Pernilla Lif Holgerson
- Department of Odontology, Section of Pediatric Dentistry, Umeå University, Umeå, Sweden.
| | - Anders Esberg
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| | - Christina E West
- Department of Clinical Sciences, Section of Pediatrics, Umeå University, Umeå, Sweden
| | - Ingegerd Johansson
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| |
Collapse
|
23
|
Gastrointestinal Tract, Microbiota and Multiple Sclerosis (MS) and the Link Between Gut Microbiota and CNS. Curr Microbiol 2023; 80:38. [DOI: 10.1007/s00284-022-03150-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
|
24
|
Bashir ST, Chiu K, Zheng E, Martinez A, Chiu J, Raj K, Stasiak S, Lai NZE, Arcanjo RB, Flaws JA, Nowak RA. Subchronic exposure to environmentally relevant concentrations of di-(2-ethylhexyl) phthalate differentially affects the colon and ileum in adult female mice. CHEMOSPHERE 2022; 309:136680. [PMID: 36209858 DOI: 10.1016/j.chemosphere.2022.136680] [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: 05/18/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a large-molecular-weight phthalate added to plastics to impart versatile properties. DEHP can be found in medical equipment and devices, food containers, building materials, and children's toys. Although DEHP exposure occurs most commonly by ingesting contaminated foods in the majority of the population, its effects on the gastrointestinal tract have not been well studied. Therefore, we analyzed the effects of subchronic exposure to DEHP on the ileum and colon morphology, gene expression, and immune microenvironment. Adult C57BL/6 female mice were orally dosed with corn oil (control, n = 7) or DEHP (0.02, 0.2, or 30 mg/kg, n = 7/treatment dose) for 30-34 days. Mice were euthanized during diestrus, and colon and ileum tissues were collected for RT-qPCR and immunohistochemistry. Subchronic DEHP exposure in the ileum altered the expression of several immune-mediating factors (Muc1, Lyz1, Cldn1) and cell viability factors (Bcl2 and Aifm1). Similarly, DEHP exposure in the colon impacted the gene expression of factors involved in mediating immune responses (Muc3a, Zo2, Ocln, Il6, and Il17a); and also altered the expression of cell viability factors (Ki67, Bcl2, Cdk4, and Aifm1) as well as a specialized epithelial cell marker (Vil1). Immunohistochemical analysis of the ileum showed DEHP increased expression of VIL1, CLDN1, and TNF and decreased number of T-cells in the villi. Histological analysis of the colon showed DEHP altered morphology and reduced cell proliferation. Moreover, in the colon, DEHP increased the expression of MUC2, MUC1, VIL1, CLDN1, and TNF. DEHP also increased the number of T-cells and Type 2 immune cells in the colon. These data suggest that subchronic DEHP exposure differentially affects the ileum and colon and alters colonic morphology and the intestinal immune microenvironment. These results have important implications for understanding the effects of DEHP on the gastrointestinal system.
Collapse
Affiliation(s)
- Shah Tauseef Bashir
- Department of Molecular and Integrative Physiology, College of Liberal Arts & Sciences, University of Illinois, Urbana, IL, USA; Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Karen Chiu
- Division of Nutritional Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA; Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA
| | - Eileen Zheng
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Angel Martinez
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Justin Chiu
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA; Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA
| | - Kishori Raj
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Sandra Stasiak
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Nastasia Zhen Ee Lai
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Rachel B Arcanjo
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Jodi A Flaws
- Division of Nutritional Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA; Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA; Carl R. Woese Institute of Genomic Biology, University of Illinois, Urbana, IL, USA
| | - Romana A Nowak
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA; Carl R. Woese Institute of Genomic Biology, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
25
|
Chen Z, Xu Q, Liu Y, Wei Y, He S, Lin W, Wang Y, Li L, Xu Y. Vancomycin-induced gut microbiota dysbiosis aggravates allergic rhinitis in mice by altered short-chain fatty acids. Front Microbiol 2022; 13:1002084. [PMID: 36439824 PMCID: PMC9687373 DOI: 10.3389/fmicb.2022.1002084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/06/2022] [Indexed: 06/11/2024] Open
Abstract
OBJECTIVE This study aims to explore how gut microbiota dysbiosis affects allergic rhinitis (AR) and whether short-chain fatty acids (SCFAs) play a role in this process. METHODS A mouse gut microbiota dysbiosis model was established by adding vancomycin to drinking water for 2 weeks before ovalbumin (OVA) sensitization. Then an OVA-alum AR mouse model was established by intraperitoneal OVA injection followed by nasal excitation. Hematoxylin and eosin (H&E) staining was performed to observe pathological changes in nasal and colon tissues of AR mice. Serum levels of total-IgE, OVA-sIgE, IL-4, IL-5, IL-10, and TGF-β1 were measured. The composition and diversity of the mouse gut microbiota were observed by 16S rDNA sequencing. Levels of SCFAs in feces were determined using SCFA-targeted metabolomics. Sodium butyrate (NaB) was added daily to mice on a low-fiber basal diet 2 weeks before the first sensitization, until the end of the study. RESULTS After gut microbiota dysbiosis, serum levels of the total IgE, OVA-sIgE, IL-4, and IL-5 in AR mice were significantly increased, compared with the control group. The composition and diversity of gut microbiota were significantly altered after gut microbiota dysbiosis, with the fecal SCFAs significantly reduced as well. The reduced bacterial genera after gut microbiota dysbiosis, such as Ruminococcus and Lactobacillus, were significantly and positively correlated with SCFAs. In contrast, the increased genera in the Van group, such as Escherichia-Shigella and Klebsiella, were significantly negatively correlated with SCFAs in feces. NaB treatment significantly reduced total-IgE, OVA-sIgE, IL-4, and IL-5 levels in serum, and inflammatory infiltration of the nasal and colon mucosa. In addition, serum levels of IL-10 and TGF-β1 increased significantly after NaB treatment. Foxp3 protein in the colon was upregulated considerably after NaB intervention. CONCLUSION Vancomycin-induced gut microbiota dysbiosis increased susceptibility and severity of AR, which is significantly related to reduced SCFA-producing bacteria, fecal SCFAs, and specific bacterial taxa. In addition, it was found that NaB alleviated low dietary fiber base-fed symptoms and immune status in AR mice.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingqing Xu
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yang Liu
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yihan Wei
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shancai He
- Department of Otorhinolaryngology, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Wei Lin
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yingge Wang
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li Li
- College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yuanteng Xu
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Allergy Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
26
|
Martins LF, Wasson DE, Hristov AN. Feeding dairy cows for improved metabolism and health. Anim Front 2022; 12:29-36. [PMID: 36268175 PMCID: PMC9564990 DOI: 10.1093/af/vfac059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Leoni F Martins
- Department of Animal Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Derek E Wasson
- Department of Animal Sciences, The Pennsylvania State University, University Park, PA, USA
| | | |
Collapse
|
27
|
Kumar A, Sakhare K, Bhattacharya D, Chattopadhyay R, Parikh P, Narayan KP, Mukherjee A. Communication in non-communicable diseases (NCDs) and role of immunomodulatory nutraceuticals in their management. Front Nutr 2022; 9:966152. [PMID: 36211513 PMCID: PMC9532975 DOI: 10.3389/fnut.2022.966152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Conveyance of pathogens between organisms causes communicable diseases. On the other hand, a non-communicable disease (NCD) was always thought to have no causative transmissible infective agents. Today, this clear distinction is increasingly getting blurred and NCDs are found to be associated with some transmissible components. The human microbiota carries a congregation of microbes, the majority and the most widely studied being bacteria in the gut. The adult human gut harbors ginormous inhabitant microbes, and the microbiome accommodates 150-fold more genes than the host genome. Microbial communities share a mutually beneficial relationship with the host, especially with respect to host physiology including digestion, immune responses, and metabolism. This review delineates the connection between environmental factors such as infections leading to gut dysbiosis and NCDs and explores the evidence regarding possible causal link between them. We also discuss the evidence regarding the value of appropriate therapeutic immunomodulatory nutritional interventions to reduce the development of such diseases. We behold such immunomodulatory effects have the potential to influence in various NCDs and restore homeostasis. We believe that the beginning of the era of microbiota-oriented personalized treatment modalities is not far away.
Collapse
Affiliation(s)
- Abhiram Kumar
- Esperer Onco Nutrition Pvt. Ltd., Mumbai, India
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | | | - Purvish Parikh
- Department of Clinical Haematology, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Kumar P. Narayan
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
- *Correspondence: Kumar P. Narayan,
| | | |
Collapse
|
28
|
Lam TJ, Mortensen K, Ye Y. Diversity and dynamics of the CRISPR-Cas systems associated with Bacteroides fragilis in human population. BMC Genomics 2022; 23:573. [PMID: 35953824 PMCID: PMC9367070 DOI: 10.1186/s12864-022-08770-8] [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: 02/15/2022] [Accepted: 07/15/2022] [Indexed: 11/22/2022] Open
Abstract
Background CRISPR-Cas (clustered regularly interspaced short palindromic repeats—CRISPR-associated proteins) systems are adaptive immune systems commonly found in prokaryotes that provide sequence-specific defense against invading mobile genetic elements (MGEs). The memory of these immunological encounters are stored in CRISPR arrays, where spacer sequences record the identity and history of past invaders. Analyzing such CRISPR arrays provide insights into the dynamics of CRISPR-Cas systems and the adaptation of their host bacteria to rapidly changing environments such as the human gut. Results In this study, we utilized 601 publicly available Bacteroides fragilis genome isolates from 12 healthy individuals, 6 of which include longitudinal observations, and 222 available B. fragilis reference genomes to update the understanding of B. fragilis CRISPR-Cas dynamics and their differential activities. Analysis of longitudinal genomic data showed that some CRISPR array structures remained relatively stable over time whereas others involved radical spacer acquisition during some periods, and diverse CRISPR arrays (associated with multiple isolates) co-existed in the same individuals with some persisted over time. Furthermore, features of CRISPR adaptation, evolution, and microdynamics were highlighted through an analysis of host-MGE network, such as modules of multiple MGEs and hosts, reflecting complex interactions between B. fragilis and its invaders mediated through the CRISPR-Cas systems. Conclusions We made available of all annotated CRISPR-Cas systems and their target MGEs, and their interaction network as a web resource at https://omics.informatics.indiana.edu/CRISPRone/Bfragilis. We anticipate it will become an important resource for studying of B. fragilis, its CRISPR-Cas systems, and its interaction with mobile genetic elements providing insights into evolutionary dynamics that may shape the species virulence and lead to its pathogenicity. Supplementary Information The online version contains supplementary material available at (10.1186/s12864-022-08770-8).
Collapse
Affiliation(s)
- Tony J Lam
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA
| | - Kate Mortensen
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA
| | - Yuzhen Ye
- School of Informatics, Computing and Engineering, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
29
|
Shen L, Zhang W, Yuan Y, Zhu W, Shang A. Vaginal microecological characteristics of women in different physiological and pathological period. Front Cell Infect Microbiol 2022; 12:959793. [PMID: 35937699 PMCID: PMC9354832 DOI: 10.3389/fcimb.2022.959793] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
The vaginal microbiota, the host endocrine system, the vaginal anatomy, and the local mucosal immunity comprise the vaginal microbiota, which interacts with each other to maintain the balance of the vaginal microbiota, which maintains female reproductive health. Puberty, menstruation, pregnancy, and menopause are four phases women go through during their reproductive and post-reproductive years. Vaginal microbiota composition and abundance are heavily influenced by estrogen and progesterone, which start at puberty and continue during the reproductive years in a dynamic balance with some fluctuations. Estrogen promotes proliferation of vaginal epithelial cells and increases glycogen storage, while progesterone lyses vaginal epithelial cells, facilitating the release of glycogen to maintain normal pH. This review summarizes the latest national and international evidence on the composition and distribution of vaginal microecology in women during different physiological and pathological periods and proposes a hormone-driven microbial diversity hypothesis to explain the temporal patterns of vaginal microbial diversity during the female reproductive cycle and menopause. A relatively balanced vaginal microecological system has a positive effect on the maintenance of female health. An imbalance in the ratio of flora can lead to susceptibility to infections or reproductive complications. The study of human microecology and its role in the development and progression of human disease is essential for the prevention, diagnosis, and treatment of related obstetric and gynecologic conditions.
Collapse
Affiliation(s)
- Liping Shen
- Department of Obstetrics and Gynecology, Changning Maternity & Infant Health Hospital, Shanghai, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Laboratory Medicine, Jiaozuo Fifth People’s Hospital, Jiaozuo, China
| | - Yi Yuan
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weipei Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Anquan Shang, ; Weipei Zhu,
| | - Anquan Shang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Anquan Shang, ; Weipei Zhu,
| |
Collapse
|
30
|
Thirunavukarasu AJ, Ross AC, Gilbert RM. Vitamin A, systemic T-cells, and the eye: Focus on degenerative retinal disease. Front Nutr 2022; 9:914457. [PMID: 35923205 PMCID: PMC9339908 DOI: 10.3389/fnut.2022.914457] [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: 04/06/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The first discovered vitamin, vitamin A, exists in a range of forms, primarily retinoids and provitamin carotenoids. The bioactive forms of vitamin A, retinol and retinoic acid, have many critical functions in body systems including the eye and immune system. Vitamin A deficiency is associated with dysfunctional immunity, and presents clinically as a characteristic ocular syndrome, xerophthalmia. The immune functions of vitamin A extend to the gut, where microbiome interactions and nutritional retinoids and carotenoids contribute to the balance of T cell differentiation, thereby determining immune status and contributing to inflammatory disease around the whole body. In the eye, degenerative conditions affecting the retina and uvea are influenced by vitamin A. Stargardt's disease (STGD1; MIM 248200) is characterised by bisretinoid deposits such as lipofuscin, produced by retinal photoreceptors as they use and recycle a vitamin A-derived chromophore. Age-related macular degeneration features comparable retinal deposits, such as drusen featuring lipofuscin accumulation; and is characterised by parainflammatory processes. We hypothesise that local parainflammatory processes secondary to lipofuscin deposition in the retina are mediated by T cells interacting with dietary vitamin A derivatives and the gut microbiome, and outline the current evidence for this. No cures exist for Stargardt's or age-related macular degeneration, but many vitamin A-based therapeutic approaches have been or are being trialled. The relationship between vitamin A's functions in systemic immunology and the eye could be further exploited, and further research may seek to leverage the interactions of the gut-eye immunological axis.
Collapse
Affiliation(s)
- Arun J. Thirunavukarasu
- Corpus Christi College, University of Cambridge, Cambridge, United Kingdom
- University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - A. Catharine Ross
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Rose M. Gilbert
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
31
|
Diamond L, Wine R, Morris SK. Impact of intrapartum antibiotics on the infant gastrointestinal microbiome: a narrative review. Arch Dis Child 2022; 107:627-634. [PMID: 34716171 DOI: 10.1136/archdischild-2021-322590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The composition of the infant gastrointestinal (GI) microbiome has been linked to adverse long-term health outcomes and neonatal sepsis. Several factors are known to impact the composition of the microbiome, including mode of delivery, gestational age, feeding method and exposure to antibiotics. The impact of intrapartum antibiotics (IPAs) on the infant microbiome requires further research. OBJECTIVE We aimed to evaluate the impact of IPAs on the infant GI microbiome. METHODS We searched Ovid MEDLINE and Embase Classic+Embase for articles in English reporting on the microbiome of infants exposed to IPAs from the date of inception to 3 January 2021. Primary outcomes included abundance and colonisation of Bifidobacterium and Lactobacillus, as well as alpha and beta diversity. RESULTS 30 papers were included in this review. In the first year of life, following exposure to IPAs, 30% (6/20) of infant cohorts displayed significantly reduced Bifidobacterium, 89% (17/19) did not display any significant differences in Lactobacillus colonisation, 21% (7/34) displayed significantly reduced alpha diversity and 35% (12/34) displayed alterations in beta diversity. Results were further stratified by delivery, gestational age (preterm or full term) and feeding method. CONCLUSIONS IPAs impact the composition of the infant GI microbiome, resulting in possible reductions Bifidobacterium and alpha diversity, and possible alterations in beta diversity. Our findings may have implications for maternal and neonatal health, including interventions to prevent reductions in health-promoting bacteria (eg, probiotics) and IPA class selection.
Collapse
Affiliation(s)
- Laura Diamond
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Wine
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaun K Morris
- Division of Infectious Diseases and Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada .,Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Xu AA, Kennedy LK, Hoffman K, White DL, Kanwal F, El-Serag HB, Petrosino JF, Jiao L. Dietary Fatty Acid Intake and the Colonic Gut Microbiota in Humans. Nutrients 2022; 14:2722. [PMID: 35807903 PMCID: PMC9268849 DOI: 10.3390/nu14132722] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/19/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
A high-fat diet has been associated with systemic diseases in humans and alterations in gut microbiota in animal studies. However, the influence of dietary fatty acid intake on gut microbiota in humans has not been well studied. In this cross-sectional study, we examined the association between intake of total fatty acids (TFAs), saturated fatty acids (SFAs), trans fatty acids (TrFAs), monounsaturated fatty acids (MUFAs), polyunsaturated fatty acids (PUFAs), n3-FAs, and n6-FAs, and the community composition and structure of the adherent colonic gut microbiota. We obtained 97 colonic biopsies from 34 participants with endoscopically normal colons. Microbial DNA was used to sequence the 16S rRNA V4 region. The DADA2 and SILVA database were used for amplicon sequence variant assignment. Dietary data were collected using the Block food frequency questionnaire. The biodiversity and the relative abundance of the bacterial taxa by higher vs. lower fat intake were compared using the Mann−Whitney test followed by multivariable negative binomial regression model. False discovery rate−adjusted p-values (q value) < 0.05 indicated statistical significance. The beta diversity of gut bacteria differed significantly by intake of all types of fatty acids. The relative abundance of Sutterella was significantly higher with higher intake of TFAs, MUFAs, PUFAs, and n6-FAs. The relative abundance of Tyzzerella and Fusobacterium was significantly higher with higher intake of SFAs. Tyzzerella was also higher with higher intake of TrFA. These observations were confirmed by multivariate analyses. Dietary fat intake was associated with bacterial composition and structure. Sutterella, Fusobacterium, and Tyzzerella were associated with fatty acid intake.
Collapse
Affiliation(s)
- Anthony A. Xu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
| | - Luke K. Kennedy
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
| | - Kristi Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (K.H.); (J.F.P.)
| | - Donna L. White
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Section of Gastroenterology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Hashem B. El-Serag
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Section of Gastroenterology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Joseph F. Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (K.H.); (J.F.P.)
| | - Li Jiao
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (A.A.X.); (L.K.K.); (D.L.W.); (F.K.); (H.B.E.-S.)
| |
Collapse
|
33
|
Zheng L, Ji YY, Wen XL, Duan SL. Fecal microbiota transplantation in the metabolic diseases: Current status and perspectives. World J Gastroenterol 2022; 28:2546-2560. [PMID: 35949351 PMCID: PMC9254144 DOI: 10.3748/wjg.v28.i23.2546] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/14/2022] [Accepted: 05/07/2022] [Indexed: 02/06/2023] Open
Abstract
With the development of microbiology and metabolomics, the relationship between the intestinal microbiome and intestinal diseases has been revealed. Fecal microbiota transplantation (FMT), as a new treatment method, can affect the course of many chronic diseases such as metabolic syndrome, malignant tumor, autoimmune disease and nervous system disease. Although the mechanism of action of FMT is now well understood, there is some controversy in metabolic diseases, so its clinical application may be limited. Microflora transplantation is recommended by clinical medical guidelines and consensus for the treatment of recurrent or refractory Clostridium difficile infection, and has been gradually promoted for the treatment of other intestinal and extraintestinal diseases. However, the initial results are varied, suggesting that the heterogeneity of the donor stools may affect the efficacy of FMT. The success of FMT depends on the microbial diversity and composition of donor feces. Therefore, clinical trials may fail due to the selection of ineffective donors, and not to faulty indication selection for FMT. A new understanding is that FMT not only improves insulin sensitivity, but may also alter the natural course of type 1 diabetes by modulating autoimmunity. In this review, we focus on the main mechanisms and deficiencies of FMT, and explore the optimal design of FMT research, especially in the field of cardiometabolic diseases.
Collapse
Affiliation(s)
- Lie Zheng
- Department of Gastroenterology, Shaanxi Hospital of Traditional Chinese Medicine, Xi’an 710003, Shaanxi Province, China
| | - Yong-Yi Ji
- Department of Neurology, Xi’an Hospital of Traditional Chinese Medicine, Xi’an 710021, Shaanxi Province, China
| | - Xin-Li Wen
- Department of Gastroenterology, Shaanxi Hospital of Traditional Chinese Medicine, Xi’an 710003, Shaanxi Province, China
| | - Sheng-Lei Duan
- Department of Gastroenterology, Shaanxi Hospital of Traditional Chinese Medicine, Xi’an 710003, Shaanxi Province, China
| |
Collapse
|
34
|
Han K, Ahn Y, Hong KB, Suh HJ, Yu KW, Kim H. Ameliorating the efficacy of galacto-oligosaccharides on ovalumin-induced allergic dermatitis symptoms in Balb/c mice by regulating Th2 immune response and the ecosystem of gut microbiota. Food Funct 2022; 13:6271-6281. [PMID: 35604005 DOI: 10.1039/d2fo00157h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, we evaluated the effect of oral administration of galacto-oligosaccharide (GOS), famous biological molecules that are comprised of galactose and lactose, on ovalbumin (OVA)-induced allergic dermatitis. OVA-induced mice were divided into the OVA-administered group (OVA-C), promethazine hydrochloride-administered group (PC), and 100 and 200 mg kg-1 GOS-administered groups (GL and GH, respectively). GOS administration significantly improved epidermal thickness and decreased CD4+ cell numbers. The concentrations of IgE, OVA-specific IgE, and inflammatory cytokines (IL-4, IL-5, and INF-γ) in GH group mice were significantly lower than those in OVA-C group mice. Firmicutes and Bacteroidetes were identified as the major phyla in the intestinal microbiota in mice, and the relative abundance of Deferribacteres was significantly lower in the GH group than in the OVA-C group. Deferribacteraceae and Mucispirillum species were significantly lower in the GH group than in the OVA-C group. The relative abundance of Muribaculum species was significantly lower, but those of Lachnospira and Lactococcus species were significantly higher in the GH group than in the OVA-C group. Our results suggest that the alleviation effect of GOS on allergic dermatitis induced by OVA sensitization was achieved by regulating hypersensitive immune responses by improving the intestinal microbial ecosystem.
Collapse
Affiliation(s)
- Kisoo Han
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Yejin Ahn
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea.
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea. .,Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Republic of Korea
| | - Kwang-Won Yu
- Division of Food and Nutrition, Korea National University of Transportation, Republic of Korea.
| | - Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
35
|
Domestic Environment and Gut Microbiota: Lessons from Pet Dogs. Microorganisms 2022; 10:microorganisms10050949. [PMID: 35630391 PMCID: PMC9143008 DOI: 10.3390/microorganisms10050949] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Accumulating data show the involvement of intestinal microbiota in the development and maintenance of numerous diseases. Many environmental factors influence the composition and function of the gut microbiota. An animal model subjected to the same environmental constraints that will allow better characterization of the microbiota–host dialogue is awaited. The domestic dog has physiological, dietary and pathological characteristics similar to those of humans and shares the domestic environment and lifestyle of its owner. This review exposes how the domestication of dogs has brought them closer to humans based on their intrinsic and extrinsic similarities which were discerned through examining and comparing the current knowledge and data on the intestinal microbiota of humans and canines in the context of several spontaneous pathologies, including inflammatory bowel disease, obesity and diabetes mellitus.
Collapse
|
36
|
Shivani S, Kao CY, Chattopadhyay A, Chen JW, Lai LC, Lin WH, Lu TP, Huang IH, Tsai MH, Teng CH, Wu JJ, Hsieh YH, Wang MC, Chuang EY. Uremic Toxin-Producing Bacteroides Species Prevail in the Gut Microbiota of Taiwanese CKD Patients: An Analysis Using the New Taiwan Microbiome Baseline. Front Cell Infect Microbiol 2022; 12:726256. [PMID: 35558102 PMCID: PMC9086402 DOI: 10.3389/fcimb.2022.726256] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 03/16/2022] [Indexed: 12/02/2022] Open
Abstract
Rationale and Objective Gut microbiota have been targeted by alternative therapies for non-communicable diseases. We examined the gut microbiota of a healthy Taiwanese population, identified various bacterial drivers in different demographics, and compared them with dialysis patients to associate kidney disease progression with changes in gut microbiota. Study Design This was a cross-sectional cohort study. Settings and Participants Fecal samples were obtained from 119 healthy Taiwanese volunteers, and 16S rRNA sequencing was done on the V3-V4 regions to identify the bacterial enterotypes. Twenty-six samples from the above cohort were compared with fecal samples from 22 peritoneal dialysis and 16 hemodialysis patients to identify species-level bacterial biomarkers in the dysbiotic gut of chronic kidney disease (CKD) patients. Results Specific bacterial species were identified pertaining to different demographics such as gender, age, BMI, physical activity, and sleeping habits. Dialysis patients had a significant difference in gut microbiome composition compared to healthy controls. The most abundant genus identified in CKD patients was Bacteroides, and at the species level hemodialysis patients showed significant abundance in B. ovatus, B. caccae, B. uniformis, and peritoneal dialysis patients showed higher abundance in Blautia producta (p ≤ 0.05) than the control group. Pathways pertaining to the production of uremic toxins were enriched in CKD patients. The abundance of the bacterial species depended on the type of dialysis treatment. Conclusion This study characterizes the healthy gut microbiome of a Taiwanese population in terms of various demographics. In a case-control examination, the results showed the alteration in gut microbiota in CKD patients corresponding to different dialysis treatments. Also, this study identified the bacterial species abundant in CKD patients and their possible role in complicating the patients’ condition.
Collapse
Affiliation(s)
- Subhashree Shivani
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Amrita Chattopadhyay
- Center for Translational Genomic Research, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jenn-Wei Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Hung Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Pin Lu
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Hsiu Huang
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Mong-Hsun Tsai
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ching-Hao Teng
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiunn-Jong Wu
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsien Hsieh
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Cheng Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- *Correspondence: Eric Y. Chuang, ; Ming-Cheng Wang,
| | - Eric Y. Chuang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
- Master Program for Biomedical Engineering, China Medical University, Taichung, Taiwan
- *Correspondence: Eric Y. Chuang, ; Ming-Cheng Wang,
| |
Collapse
|
37
|
Korsten SGPJ, Peracic L, van Groeningen LMB, Diks MAP, Vromans H, Garssen J, Willemsen LEM. Butyrate Prevents Induction of CXCL10 and Non-Canonical IRF9 Expression by Activated Human Intestinal Epithelial Cells via HDAC Inhibition. Int J Mol Sci 2022; 23:ijms23073980. [PMID: 35409339 PMCID: PMC8999521 DOI: 10.3390/ijms23073980] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
Non-communicable diseases are increasing and have an underlying low-grade inflammation in common, which may affect gut health. To maintain intestinal homeostasis, unwanted epithelial activation needs to be avoided. This study compared the efficacy of butyrate, propionate and acetate to suppress IFN-γ+/−TNF-α induced intestinal epithelial activation in association with their HDAC inhibitory capacity, while studying the canonical and non-canonical STAT1 pathway. HT-29 were activated with IFN-γ+/−TNF-α and treated with short chain fatty acids (SCFAs) or histone deacetylase (HDAC) inhibitors. CXCL10 release and protein and mRNA expression of proteins involved in the STAT1 pathway were determined. All SCFAs dose-dependently inhibited CXCL10 release of the cells after activation with IFN-γ or IFN-γ+TNF-α. Butyrate was the most effective, completely preventing CXCL10 induction. Butyrate did not affect phosphorylated STAT1, nor phosphorylated NFκB p65, but inhibited IRF9 and phosphorylated JAK2 protein expression in activated cells. Additionally, butyrate inhibited CXCL10, SOCS1, JAK2 and IRF9 mRNA in activated cells. The effect of butyrate was mimicked by class I HDAC inhibitors and a general HDAC inhibitor Trichostatin A. Butyrate is the most potent inhibitor of CXCL10 release compared to other SCFAs and acts via HDAC inhibition. This causes downregulation of CXCL10, JAK2 and IRF9 genes, resulting in a decreased IRF9 protein expression which inhibits the non-canonical pathway and CXCL10 transcription.
Collapse
Affiliation(s)
- Sandra G. P. J. Korsten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands;
- Correspondence: (S.G.P.J.K.); (L.E.M.W.)
| | - Laura Peracic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Luka M. B. van Groeningen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Mara A. P. Diks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Herman Vromans
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands;
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Nutricia Research B.V., 3584 CT Utrecht, The Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Correspondence: (S.G.P.J.K.); (L.E.M.W.)
| |
Collapse
|
38
|
Cataldi S, Bonavolontà V, Poli L, Clemente FM, De Candia M, Carvutto R, Silva AF, Badicu G, Greco G, Fischetti F. The Relationship between Physical Activity, Physical Exercise, and Human Gut Microbiota in Healthy and Unhealthy Subjects: A Systematic Review. BIOLOGY 2022; 11:479. [PMID: 35336852 PMCID: PMC8945171 DOI: 10.3390/biology11030479] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023]
Abstract
Several studies have been conducted to find at least an association between physical activity (PA)/ physical exercise (PE) and the possibility to modulate the gut microbiome (GM). However, the specific effects produced on the human GM by different types of PA/PE, different training modalities, and their age-related effects are not yet fully understood. Therefore, this systematic review aims to evaluate and summarize the current scientific evidence investigating the bi-directional relationship between PA/PE and the human GM, with a specific focus on the different types/variables of PA/PE and age-related effects, in healthy and unhealthy people. A systematic search was conducted across four databases (Web of Science, Medline (PubMed), Google Scholar, and Cochrane Library). Information was extracted using the populations, exposure, intervention, comparison, outcomes (PICOS) format. The Oxford Quality Scoring System Scale, the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I) tool, and the JBI Critical Appraisal Checklist for Analytical Cross-Sectional Studies were used as a qualitative measure of the review. The protocol was registered in PROSPERO (code: CRD42022302725). The following data items were extracted: author, year of publication, study design, number and age of participants, type of PA/PE carried out, protocol/workload and diet assessment, duration of intervention, measurement tools used, and main outcomes. Two team authors reviewed 694 abstracts for inclusion and at the end of the screening process, only 76 full texts were analyzed. Lastly, only 25 research articles met the eligibility criteria. The synthesis of these findings suggests that GM diversity is associated with aerobic exercise contrary to resistance training; abundance of Prevotella genus seems to be correlated with training duration; no significant change in GM richness and diversity are detected when exercising according to the minimum dose recommended by the World Health Organizations; intense and prolonged PE can induce a higher abundance of pro-inflammatory bacteria; PA does not lead to significant GM α/β-diversity in elderly people (60+ years). The heterogeneity of the training parameters used in the studies, diet control, and different sequencing methods are the main confounders. Thus, this systematic review can provide an in-depth overview of the relationship between PA/PE and the human intestinal microbiota and, at the same time, provide indications from the athletic and health perspective.
Collapse
Affiliation(s)
- Stefania Cataldi
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Valerio Bonavolontà
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Luca Poli
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Filipe Manuel Clemente
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal; (F.M.C.); (A.F.S.)
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), 4960-320 Melgaço, Portugal
- Instituto de Telecomunicações, Delegação da Covilhã, 1049-001 Lisboa, Portugal
| | - Michele De Candia
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Roberto Carvutto
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Ana Filipa Silva
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal; (F.M.C.); (A.F.S.)
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), 4960-320 Melgaço, Portugal
- The Research Centre in Sports Sciences, Health Sciences and Human Development (CIDESD), 5001-801 Vila Real, Portugal
| | - Georgian Badicu
- Department of Physical Education and Special Motricity, Transilvania University of Brasov, 500068 Brasov, Romania;
| | - Gianpiero Greco
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Francesco Fischetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| |
Collapse
|
39
|
Mir RA, Nazir M, Naik S, Mukhtar S, Ganai BA, Zargar SM. Utilizing the underutilized plant resources for development of life style foods: Putting nutrigenomics to use. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2022; 171:128-138. [PMID: 34998100 DOI: 10.1016/j.plaphy.2021.12.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/26/2021] [Accepted: 12/30/2021] [Indexed: 06/14/2023]
Abstract
Sufficient amount of minerals, vitamins, and proteins in human diet play indispensable role in maintaining the active metabolism for better human health. All the essential nutrients that are requisite for an individual's survival are acquired from plants as well as animals. Micronutrients and macronutrients directly influence the metabolic pathways and their deficiencies play a substantial role in development of manifold disorders. In addition to environmental factors, quality and quantity of foods are key factors in maintaining the human health. Transition from healthy to diseased state is concurrent with the pattern of gene expression that is largely influenced by nutrition and environment. A combined approach to study the influence of nutrition on expression of numerous genes can be well explored through nutrigenomic studies. Nutrigenomics includes studies wherein applied genomics is used to investigate nutritional science to understand the compartmentalization of genes that influence the cause of diet-related complications. This review describes the role of underutilized crops as frontline foods to circumvent the health complications through the nutrigenomic studies. Further dynamics of nutrigenomic tools to study the impact of nutrition on the changing pattern of genome stability and gene expression for developing precise safety measures against wide range of health ailments linked to metabolic networks. Additionally, this review provides detailed information on nutrigenomic studies undertaken to unravel the potential of underutilized crops to augment the human health and to carry the agronomic/genomic approaches to enhance nutritional profile of underutilized crops to overcome diet-related disorders.
Collapse
Affiliation(s)
- Rakeeb Ahmad Mir
- Department of Biotechnology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185131, India
| | - Muslima Nazir
- Centre of Research for Development, University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Samiullah Naik
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India
| | - Shazia Mukhtar
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development, University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Sajad Majeed Zargar
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India.
| |
Collapse
|
40
|
Ahn JR, Lee SH, Kim B, Nam MH, Ahn YK, Park YM, Jeong SM, Park MJ, Song KB, Lee SY, Hong SJ. Ruminococcus gnavus ameliorates atopic dermatitis by enhancing Treg cell and metabolites in BALB/c mice. Pediatr Allergy Immunol 2022; 33:e13678. [PMID: 34633714 DOI: 10.1111/pai.13678] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ruminococcus gnavus (R. gnavus) are mucin-degrading gut bacteria that play a key role in the early colonization of the gut by serving as endogenous sources of nutrients. They can also influence immune development. We had previously reported a lower abundance of R. gnavus in infants with atopic dermatitis (AD) compared with that in healthy subjects. However, the underlying mechanisms remain unclear. In this study, we investigated the effect of orally administered R. gnavus on antibiotic treatment-induced gut dysbiosis (and the underlying mechanism) in a mouse model of AD. METHODS Four-week-old female BALB/C mice were administered antibiotic cocktails for 2 weeks. R. gnavus was orally administered throughout the study duration. At 6 weeks of age, AD was induced by epidermal sensitization with ovalbumin. AD phenotypes and systemic and gut immune responses were investigated. RESULTS Orally administered R. gnavus significantly reduced AD-associated parameters (i.e., transepidermal water loss, clinical score, total serum immunoglobulin (Ig) E level, OVA-specific IgE level, and skin inflammation). R. gnavus treatment also resulted in significant downregulation of T helper 2-related cytokine mRNA and upregulation of interleukin (IL)-10 and Foxp3 in the skin. The population of CD4+ FOXP3+ T cells in mesenteric- and skin-draining lymph nodes and butyrate levels in the cecum increased in R. gnavus-administered AD mice. CONCLUSIONS Immune modulation by orally administered R. gnavus may alleviate AD symptoms through the enhancement of regulatory T-cell counts and short-chain fatty acids production in AD mice.
Collapse
Affiliation(s)
- Jae-Rin Ahn
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Hwa Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Byunghyun Kim
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Myung Hee Nam
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Yoon Kyung Ahn
- Korea Basic Science Institute, Western Seoul Center, Seoul, Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seon-Mi Jeong
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Jee Park
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kun Baek Song
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Trained immunity in type 2 immune responses. Mucosal Immunol 2022; 15:1158-1169. [PMID: 36065058 PMCID: PMC9705254 DOI: 10.1038/s41385-022-00557-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
Immunological memory of innate immune cells, also termed "trained immunity", allows for cross-protection against distinct pathogens, but may also drive chronic inflammation. Recent studies have shown that memory responses associated with type 2 immunity do not solely rely on adaptive immune cells, such as T- and B cells, but also involve the innate immune system and epithelial cells. Memory responses have been described for monocytes, macrophages and airway epithelial cells of asthmatic patients as well as for macrophages and group 2 innate lymphoid cells (ILC2) from allergen-sensitized or helminth-infected mice. The metabolic and epigenetic mechanisms that mediate allergen- or helminth-induced reprogramming of innate immune cells are only beginning to be uncovered. Trained immunity has been implicated in helminth-driven immune regulation and allergen-specific immunotherapy, suggesting its exploitation in future therapies. Here, we discuss recent advances and key remaining questions regarding the mechanisms and functions of trained type 2 immunity in infection and inflammation.
Collapse
|
42
|
Sirufo MM, De Pietro F, Catalogna A, Ginaldi L, De Martinis M. The Microbiota-Bone-Allergy Interplay. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:ijerph19010282. [PMID: 35010543 PMCID: PMC8750778 DOI: 10.3390/ijerph19010282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022]
Abstract
Emerging knowledge suggests an increasing importance of gut microbiota in health and disease. Allergy and bone metabolism are closely interconnected, and the possible negative effects of common therapies are not the only aspects of this relationship. The immune system is influenced by the microbiota-host interactions, and several pieces of evidence suggest the existence of an interplay between microbiota, bone metabolism, and allergies. Understanding these inter-relationships is essential for the development of new potential strategies of treatment and prevention targeting microbiota. A wide range of substances and germs, prebiotics and probiotics, are capable of influencing and modifying the microbiota. Prebiotics and probiotics have been shown in several studies to have different actions based on various factors such as sex, hormonal status, and age. In this review, we summarize the latest knowledge on the topic, and we discuss practical implications and the need for further studies.
Collapse
Affiliation(s)
- Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi n. 1, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (A.C.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04, 64100 Teramo, Italy
| | - Francesca De Pietro
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi n. 1, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (A.C.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04, 64100 Teramo, Italy
| | - Alessandra Catalogna
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi n. 1, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (A.C.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04, 64100 Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi n. 1, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (A.C.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04, 64100 Teramo, Italy
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi n. 1, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (A.C.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04, 64100 Teramo, Italy
- Correspondence: ; Tel.: +39-0861-429548
| |
Collapse
|
43
|
Morales JS, Valenzuela PL, Castillo-García A, Butragueño J, Jiménez-Pavón D, Carrera-Bastos P, Lucia A. The Exposome and Immune Health in Times of the COVID-19 Pandemic. Nutrients 2021; 14:24. [PMID: 35010900 PMCID: PMC8746533 DOI: 10.3390/nu14010024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the importance of lifestyle and environmental exposures-collectively referred to as the 'exposome'-for ensuring immune health. In this narrative review, we summarize and discuss the effects of the different exposome components (physical activity, body weight management, diet, sun exposure, stress, sleep and circadian rhythms, pollution, smoking, and gut microbiome) on immune function and inflammation, particularly in the context of the current coronavirus disease 2019 (COVID-19) pandemic. We highlight the potential role of 'exposome improvements' in the prevention-or amelioration, once established-of this disease as well as their effect on the response to vaccination. In light of the existing evidence, the promotion of a healthy exposome should be a cornerstone in the prevention and management of the COVID-19 pandemic and other eventual pandemics.
Collapse
Affiliation(s)
- Javier S. Morales
- MOVE-IT Research Group, Department of Physical Education, Faculty of Education Sciences, Universidad de Cádiz, 11519 Cadiz, Spain;
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, 11009 Cadiz, Spain
| | - Pedro L. Valenzuela
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (P.L.V.); (A.L.)
- Physical Activity and Health Research Group (‘PaHerg’), Research Institute of the Hospital 12 de Octubre (‘imas12′), 28041 Madrid, Spain
| | | | - Javier Butragueño
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Polytechnic University of Madrid (UPM), 28040 Madrid, Spain;
| | - David Jiménez-Pavón
- MOVE-IT Research Group, Department of Physical Education, Faculty of Education Sciences, Universidad de Cádiz, 11519 Cadiz, Spain;
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, 11009 Cadiz, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| | - Pedro Carrera-Bastos
- Centre for Primary Health Care Research, Lund University, Skane University Hospital, 205 02 Malmö, Sweden;
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (P.L.V.); (A.L.)
- Physical Activity and Health Research Group (‘PaHerg’), Research Institute of the Hospital 12 de Octubre (‘imas12′), 28041 Madrid, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| |
Collapse
|
44
|
Turta O, Selma-Royo M, Kumar H, Collado MC, Isolauri E, Salminen S, Rautava S. Maternal Intrapartum Antibiotic Treatment and Gut Microbiota Development in Healthy Term Infants. Neonatology 2021; 119:93-102. [PMID: 34808634 DOI: 10.1159/000519574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/08/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of the study was to investigate the impact of intrapartum antibiotic treatment (IAT) on the compositional development of gut microbiota in healthy term infants. STUDY DESIGN A case-control study of 24 infants exposed to and 24 matched infants not exposed to IAT was conducted. All subjects were born by vaginal delivery at term and breastfed. None of the infants received antibiotics during the immediate neonatal period. Fecal samples were obtained at the ages of 1 and 6 months. The composition of the intestinal microbiota was assessed by 16S rRNA gene sequencing. RESULTS IAT was associated with reduced microbial richness but not diversity at 1 month of age. Furthermore, the relative abundances of Clostridiaceae and Erysipelotrichaceae were significantly altered in infants exposed to IAT as compared to nonexposed infants at 1 month of age. The observed deviations in gut microbiota composition between infants exposed and not exposed to IAT diminished by the age of 6 months. CONCLUSIONS IAT is associated with short-term perturbations in the gut microbiota development in healthy term, vaginally delivered, breastfed infants. The composition of the gut microbiota is mostly restored by the age of 6 months.
Collapse
Affiliation(s)
- Olli Turta
- Department of Pediatrics, University of Turku, Turku, Finland
- Turku University Hospital, Turku, Finland
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Himanshu Kumar
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Erika Isolauri
- Department of Pediatrics, University of Turku, Turku, Finland
- Turku University Hospital, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Samuli Rautava
- Department of Pediatrics, University of Turku, Turku, Finland
| |
Collapse
|
45
|
Du L, Lei X, Wang J, Wang L, Zhong Q, Fang X, Li P, Du B, Wang Y, Liao Z. Lipopolysaccharides derived from gram-negative bacterial pool of human gut microbiota promote inflammation and obesity development. Int Rev Immunol 2021; 41:45-56. [PMID: 34726960 DOI: 10.1080/08830185.2021.1996573] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lipopolysaccharide (LPS) is the major component of the outer membrane of Gram-negative bacteria. It is found from intestinal microbes in the circulatory system and considered a trigger factor for low-grade inflammation in obesity. High-fat diet intake and its related obesity can cause gut microbiota disorder, leading to increased gut permeability, paracellular absorption and transcellular transport of endogenous endotoxin in the cardiovascular system. High-fat diet intake can also increase plasma LPS levels, and causing chronic or "low-grade" inflammation. In this review article, we summarize the recent research advancements on the mechanism of low-grade inflammation and its related obesity. We also propose several approaches that can be used to reduce endogenous endotoxin absorption.Supplemental data for this article is available online at https://doi.org/10.1080/08830185.2021.1996573 .
Collapse
Affiliation(s)
- Liyu Du
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Xi Lei
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jie Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Li Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Qingping Zhong
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yutao Wang
- College of Life and Geographic Sciences, Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, Kashi University, Kashi, China
| | - Zhenlin Liao
- College of Food Science, South China Agricultural University, Guangzhou, China.,College of Life and Geographic Sciences, Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, Kashi University, Kashi, China
| |
Collapse
|
46
|
Bunne J, Hedman L, Perzanowski M, Bjerg A, Winberg A, Andersson M, Lundbäck B, Platts-Mills T, Rönmark E. The majority of children sensitized before school-age develop allergic disease before adulthood: a longitudinal population-based study. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 10:577-585.e3. [PMID: 34695595 DOI: 10.1016/j.jaip.2021.10.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/17/2021] [Accepted: 10/06/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Allergic sensitization increases the risk of and asthma and allergic rhinitis, but the impact of age at onset of sensitization is less studied. OBJECTIVE To examine the cumulative incidence of asthma and rhinitis up to age 19 years in relation to age at onset of sensitization to airborne allergens. METHOD All children in grade one and two (median age 8 years) in two municipalities in Northern Sweden were invited to skin prick tests and a questionnaire about allergic diseases, and 88% participated. At ages 12 and 19 years the protocol was repeated, and n=1510 individuals participated in all three examinations. Specific IgE was collected in a random sample at age 19, n=770. Onset of sensitization was defined; ≤8 years, 8-12 years, 12-19 years, and never sensitized. Adjusted Poisson regression was utilized to calculate risk ratios. RESULTS At age 19, those sensitized ≤8 year had the highest risk of asthma (RR 4.68 (95%CI 3.14-6.97)) and rhinitis (RR 22.3 (95%CI 13.3-37.6)), and 84% had developed either asthma or rhinitis. The combination of sensitization ≤8 year and family history of allergic diseases rendered high risks for asthma (RR 10.6 (6.71-16.7)) and rhinitis (RR 36.3 (18.9-69.7). Individuals sensitized ≤8 year showed significantly highest level of sensitization, as judged by number of positive skin tests and titers of specific IgE. CONCLUSION The majority of individuals with sensitization ≤8 years developed asthma or rhinitis before young adulthood. The high level of sensitization in early sensitized contribute to the high incidence of allergic airway conditions.
Collapse
Affiliation(s)
- Joakim Bunne
- Department of Public Health and Clinical Medicine, Division of Sustainable Health, the OLIN unit, Umeå University, Umeå, Sweden
| | - Linnea Hedman
- Department of Public Health and Clinical Medicine, Division of Sustainable Health, the OLIN unit, Umeå University, Umeå, Sweden
| | - Matthew Perzanowski
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Anders Bjerg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Anna Winberg
- Department of Clinical Sciences, Pediatrics, Umeå University, Sweden
| | - Martin Andersson
- Department of Public Health and Clinical Medicine, Division of Sustainable Health, the OLIN unit, Umeå University, Umeå, Sweden
| | - Bo Lundbäck
- Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Platts-Mills
- Division of Allergy & Clinical Immunology, University of Virginia, Charlottesville, VA, USA
| | - Eva Rönmark
- Department of Public Health and Clinical Medicine, Division of Sustainable Health, the OLIN unit, Umeå University, Umeå, Sweden.
| |
Collapse
|
47
|
Xu AA, Hoffman K, Gurwara S, White DL, Kanwal F, El-Serag HB, Petrosino JF, Jiao L. Oral Health and the Altered Colonic Mucosa-Associated Gut Microbiota. Dig Dis Sci 2021; 66:2981-2991. [PMID: 32974807 PMCID: PMC7987909 DOI: 10.1007/s10620-020-06612-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Systemic diseases have been associated with oral health and gut microbiota. We examined the association between oral health and the community composition and structure of the adherent colonic gut microbiota. METHODS We obtained 197 snap-frozen colonic biopsies from 62 colonoscopy-confirmed polyp-free individuals. Microbial DNA was sequenced for the 16S rRNA V4 region using the Illumina MiSeq, and the sequences were assigned to the operational taxonomic unit based on SILVA. We used a questionnaire to ascertain tooth loss, gum disease, and lifestyle factors. We compared biodiversity and relative abundance of bacterial taxa based on the amount of tooth loss and the presence of gum disease. The multivariable negative binomial regression model for panel data was used to estimate the association between the bacterial count and oral health. False discovery rate-adjusted P value (q value) < .05 indicated statistical significance. RESULTS More tooth loss and gum disease were associated with lower bacterial alpha diversity. The relative abundance of Faecalibacterium was lower (q values < .05) with more tooth loss. The association was significant after adjusting for age, ethnicity, obesity, smoking, alcohol use, hypertension, diabetes, and the colon segment. The relative abundance of Bacteroides was higher in those with gum disease. CONCLUSIONS Oral health was associated with alteration in the community composition and structure of the adherent gut bacteria in the colon. The reduced anti-inflammatory Faecalibacterium in participants with more tooth loss may indicate systemic inflammation. Future studies are warranted to confirm our findings and investigate the systemic role of Faecalibacterium.
Collapse
Affiliation(s)
- Anthony A Xu
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Kristi Hoffman
- Department of Molecular Virology and Microbiology, The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Shawn Gurwara
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Donna L White
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Texas Medical Center Digestive Disease Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Section of Gastroenterology, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Texas Medical Center Digestive Disease Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Hashem B El-Serag
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Section of Gastroenterology, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Texas Medical Center Digestive Disease Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Joseph F Petrosino
- Department of Molecular Virology and Microbiology, The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Texas Medical Center Digestive Disease Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA
| | - Li Jiao
- Department of Medicine, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
- Section of Gastroenterology, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
- Texas Medical Center Digestive Disease Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd, MS152, Houston, 77030, TX, USA.
| |
Collapse
|
48
|
Wang Z, Lai Z, Zhang X, Huang P, Xie J, Jiang Q, Zhang Q, Chung KF. Altered gut microbiome compositions are associated with the severity of asthma. J Thorac Dis 2021; 13:4322-4338. [PMID: 34422359 PMCID: PMC8339736 DOI: 10.21037/jtd-20-2189] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 05/31/2021] [Indexed: 12/31/2022]
Abstract
Background Despite substantial evidence on the contribution of the diversity of the gut microbiome to the pathogenesis of asthma and allergic diseases, little is known about their relationship with asthma severity and/or clinical phenotypes. We analyzed the difference in composition of the gut microbiome between subjects with asthma and healthy subjects and explored its role in the development of asthma. Methods Fecal samples from 15 subjects with severe asthma (SA), 14 with non-severe asthma (NSA), and 15 healthy subjects were assessed by 16S ribosomal RNA gene sequencing methods to identify the gut bacterial composition. Results Compared with those in the NSA group, patients in the SA group had a higher dose of inhaled corticosteroids, and there were more atopic subjects (60% vs. 35.7%, respectively). No significant differences were found at the phylum level either in operational taxonomic unit numbers or in diversity scores among the SA, NSA, and healthy groups. However, at the family level, the relative abundance of Acidaminococcaceae in the SA group was remarkedly lower than that in the group with healthy subjects (P<0.05). Furthermore, Veillonellaceae and Prevotellaceae were significantly more common in samples from the SA group than in those from the NSA group (P<0.05). In the SA group, positive correlations were observed between the relative abundance of Veillonellaceae and mid-expiratory flow 25% (MEF25%) predicted (r=0.538, P=0.047), as well as between the relative abundance of Acidaminococcaceae and body mass index (r=0642, P=0.010). Principal component analysis suggested that the relative abundances of Acidaminococcaceae and Prevotellaceae were associated with severe asthma. Moreover, we found that class Betaproteobacteria, order Burkholderiales, and family Alcaligenaceae were significantly different among the groups defined by serum immunoglobulin E (IgE) levels. Conclusions Our findings suggest that altered gut microbiome compositions are involved in the severity of asthma and that there are specific bacteria related to different asthma phenotypes in terms of serum IgE levels.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Hexian Memorial Hospital of PanYu District, Guangzhou, China
| | - Zhengdao Lai
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Dongguan Institute of Respiratory and Critical Care Medicine, Afliated Dongguan People's Hospital, Southern Medicial University, Dongguan, China
| | - Xiaoxian Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peikai Huang
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Respiration Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Jiaxing Xie
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Jiang
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Insitute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kian Fan Chung
- National Heart & Lung Institute, Imperial College London & Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| |
Collapse
|
49
|
Prescott SL. A world of inflammation: the need for ecological solutions that co-benefit people, place and planet. Vet Dermatol 2021; 32:539-e149. [PMID: 34415086 DOI: 10.1111/vde.13013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 05/11/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
The ecology of the early environment - including microbial diversity, nutrition, nature, social interactions and the totality of exposures in the wider "exposome" - have life-long implications for all aspects of health and resilience. In particular, the emergence of "microbiome science" provides new evidence for vital relationships between biodiversity and health at every level. New perspectives of ecological interdependence connect personal and planetary health; the human health crisis cannot be separated from the social, political and economic "ecosystems" otherwise driving dysbiosis (from its etymological root, "life in distress") at every level. Adverse changes in macroscale ecology - of food systems, lifestyle behaviours, socioeconomic disadvantage and environmental degradation - all impact the microbial systems sitting at the foundations of all ecosystems. In particular, changes in the function and composition of the human-associated microbiome have been implicated in the mounting global burden of noncommunicable diseases (NCDs), exacerbating inflammation and metabolic dysregulation through multiple pathways across the lifespan. This "dysbiotic drift" (adverse shifts in ecology at all scales) underscores the need for ecological approaches aimed at restoring symbiosis, balance and mutualism. While there is promise with supplement-based strategies (e.g. probiotics, prebiotics), it is essential to focus on upstream factors implicated in dysbiosis, including the health of wider environments, lifestyle, nature relatedness, and the social policies and practices which can facilitate or inhibit dysbiotic drift. This also calls for ambitious integrative approaches which not only define these interconnections, but also capitalize on them to create novel, collaborative and mutualistic solutions to our vast interdependent global challenges.
Collapse
Affiliation(s)
- Susan L Prescott
- The ORIGINS Project, Telethon Kids Institute, Perth Children's Hospital, University of Western Australia, Nedlands, WA, 6009, Australia.,InVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, 10704, USA.,The NOVA Institute, 1407 Fleet Street, Baltimore, MD, 21231, USA.,Center for Integrative Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
50
|
Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr 2021; 12:1239-1285. [PMID: 33693453 PMCID: PMC8321864 DOI: 10.1093/advances/nmaa181] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the gut microbiota has emerged as a key component in regulating brain processes and behavior. Diet is one of the major factors involved in shaping the gut microbiota composition across the lifespan. However, whether and how diet can affect the brain via its effects on the microbiota is only now beginning to receive attention. Several mechanisms for gut-to-brain communication have been identified, including microbial metabolites, immune, neuronal, and metabolic pathways, some of which could be prone to dietary modulation. Animal studies investigating the potential of nutritional interventions on the microbiota-gut-brain axis have led to advancements in our understanding of the role of diet in this bidirectional communication. In this review, we summarize the current state of the literature triangulating diet, microbiota, and host behavior/brain processes and discuss potential underlying mechanisms. Additionally, determinants of the responsiveness to a dietary intervention and evidence for the microbiota as an underlying modulator of the effect of diet on brain health are outlined. In particular, we emphasize the understudied use of whole-dietary approaches in this endeavor and the need for greater evidence from clinical populations. While promising results are reported, additional data, specifically from clinical cohorts, are required to provide evidence-based recommendations for the development of microbiota-targeted, whole-dietary strategies to improve brain and mental health.
Collapse
Affiliation(s)
| | | | - Wolfgang Marx
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
| | - Harriet Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Felice Jacka
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Black Dog Institute, Randwick, NSW, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Timothy G Dinan
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|