1
|
Miyoshi J, Hisamatsu T. Effect of maternal exposure to antibiotics during pregnancy on the neonatal intestinal microbiome and health. Clin J Gastroenterol 2025; 18:1-10. [PMID: 39709577 DOI: 10.1007/s12328-024-02088-6] [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: 08/16/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Antibiotics are widely used during pregnancy. Recent epidemiological studies suggest that maternal exposure to antibiotics during pregnancy is associated with increased risks of various diseases in offspring; host-microbiome interactions are considered to be involved in pathogenesis, as antibiotic-induced perturbations (dysbiosis) of the maternal microbiome can be transmitted to offspring. We reviewed the current status of antibiotic usage during pregnancy, transmission of maternal antibiotic-induced dysbiosis to offspring, and several diseases in offspring reported to be associated with maternal antibiotic exposure. Antibiotics must be properly used when necessary. While the adverse effect of maternal antibiotic exposure during pregnancy on the health of offspring has been demonstrated by several studies, more robust clinical evidence is necessary to define the best practice for antibiotic use during pregnancy. Epidemiologic studies have limitations in establishing causal links beyond associations; animal studies provide benefits in examining these links, however, microbiomes, gestation courses, and aging vary between host species. Understanding the underlying mechanisms of epidemiologic findings as well as the healthy microbiome during pregnancy and early life in humans would contribute to developing future microbial interventions for restoring antibiotic-induced dysbiosis during pregnancy.
Collapse
Affiliation(s)
- Jun Miyoshi
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan.
| | - Tadakazu Hisamatsu
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| |
Collapse
|
2
|
Scharf E, Schlattmann P, Stallhofer J, Stallmach A. Do Antibiotics Cause Inflammatory Bowel Disease? A Systematic Review and Meta-Analysis. Visc Med 2025; 41:32-47. [PMID: 39927188 PMCID: PMC11801854 DOI: 10.1159/000541601] [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: 05/18/2024] [Accepted: 09/23/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), exhibits a multifactorial pathogenesis influenced by genetic and environmental factors. Antibiotic usage has been implicated in modifying the gut microbiome, potentially leading to dysbiosis and contributing to IBD risk. Despite existing literature, the relationship remains inconclusive. This meta-analysis aimed to evaluate the association between prior antibiotic use and the onset of IBD. Methods A systematic literature search in PubMed was conducted to identify studies exploring the link between antibiotic use and subsequent IBD diagnosis. Studies reporting CD, UC, or both as primary outcomes were included. The meta-analysis, performed according to PRISMA guidelines, summarized risk estimates, represented as odds ratios (ORs), and corresponding confidence intervals (CIs). Subgroup analyses involved the categorization of antibiotics and the determination of the minimum number of antibiotic therapy courses administered. Results Out of 722 publications, 31 studies comprising 102,103 individuals met eligibility criteria. The pooled OR for IBD in those with prior antibiotic exposure was 1.40 (95% CI: 1.25-1.56). Antibiotic use was associated with an increased risk of IBD (OR: 1.52, 95% CI: 1.19-1.94). Notably, this association was confined to CD (OR: 1.50, 95% CI: 1.27-1.77), while no significant association was observed with UC (OR: 1.21, 95% CI: 1.00-1.47). Risk augmentation for IBD correlated positively with the number of antibiotic courses (OR: 1.08, 95% CI: 1.05-1.12). Conclusion Previous antibiotic use is associated with the later development of CD. A positive dose-response effect was also observed. Against this background, antibiotics should be used rationally.
Collapse
Affiliation(s)
- Ellen Scharf
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Peter Schlattmann
- Institute for Medical Statistics, Informatics, and Data Science, Jena University Hospital, Jena, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| |
Collapse
|
3
|
Islam MF, Arka PB, Rohman M, Hossain MS, Babu MR, Azhari HA, Uddin MJ. Pooling the complex survey data across the 64 lower and middle-income countries: A study on antibiotic usage in under-five children. Heliyon 2025; 11:e41470. [PMID: 39834425 PMCID: PMC11743117 DOI: 10.1016/j.heliyon.2024.e41470] [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: 04/24/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
Background Antibiotic exposure for fever/cough has been rising alarmingly, especially among children under five in low- and middle-income countries (LMICs). This excessive use could result in higher healthcare costs, and antibiotic resistance is an alarming trend in developing countries. As a result, it's crucial to look at the variables that affect antibiotic exposure and highlight the subgroups among whom antibiotic abuse is the most prevalent. Methods We used the most recent standard Demographic and Health Survey (DHS) data for 64 LMICs. Sample weights were employed in studies to ensure exact standard errors and estimate p-values. To analyze antibiotic exposures across countries, continents, economic levels, and the top and bottom ten countries with fever/cough, we conducted descriptive statistics. Additionally, we provided accompanying bar diagrams for each descriptive finding to enhance visual understanding. For geospatial analysis, we utilized ArcGIS, a powerful tool for mapping and spatial analysis. Findings We used data from 141,018 children under five who were reported to have had fever/cough recently in 64 LMICs. Among them, 30.4 % were exposed to antibiotics for fever/cough. Congo (68.7 %), Egypt (65.5 %), and Tajikistan (61.8 %) had the highest prevalence of antibiotic consumption across LMICs. On the other hand, Cameroon (0.3 %), Armenia (4.0 %), and Mauritania (6.1 %) had the lowest. In the final binary logistic regression, mothers that had any formal education (Primary: OR = 1.28, 95 % CI:1.07-1.53, Secondary: OR = 1.38, 95 % CI:1.14-1.67, Higher Education: OR = 1.69, 95 % CI:1.23-2.33) were more likely to expose their children to antibiotics for fever/cough than illiterate ones. Additionally, children in richer households (Richer: OR = 1.38, 95 % CI:1.15-1.66, Richest: OR = 1.46, 95 % CI:1.19-1.80) were more likely to receive antibiotics than those in poorer households. Conclusions Antibiotic exposure for fever/cough in children under five was relatively moderate across 64 LMICs. However, the rate was incredibly high in some LMICs and incredibly low in others. The researchers recommend that the countries with high and low percentages of antibiotic exposure investigate either any possibility of antibiotic abuse or proper healthcare service at the national level.
Collapse
Affiliation(s)
- Md Fakrul Islam
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Prosenjit Basak Arka
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahfuzer Rohman
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Md Sabbir Hossain
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Md Rashed Babu
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Hasin Anupama Azhari
- Institute of Natural Sciences, United International University, Dhaka, Bangladesh
| | - Md Jamal Uddin
- Biostatistics, Epidemiology and Public Health Research Team, Department of Statistics, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
- Faculty of Graduate Studies, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
4
|
Mårild K, Lerchova T, Östensson M, Imberg H, Størdal K, Ludvigsson J. Early-Life Infections, Antibiotics and Later Risk of Childhood and Early Adult-Onset Inflammatory Bowel Disease: Pooled Analysis of Two Scandinavian Birth Cohorts. Aliment Pharmacol Ther 2025; 61:323-334. [PMID: 39450871 DOI: 10.1111/apt.18358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/31/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Childhood antibiotic use has been associated with inflammatory bowel disease (IBD), although the potential contribution of infection frequency remains uncertain. AIMS To explore the association between early-life infections, antibiotics and IBD development. METHODS We used population-based data from ABIS (Sweden) and MoBa (Norway) cohorts following children from birth (1997-2009) until 2021. Prospectively collected questionnaires identified infection frequency (any, gastrointestinal and respiratory) and antibiotics (any, penicillin and non-penicillin) until age 3. IBD diagnosis required ≥ 2 records in national health registries. Cohort-specific hazard ratios (aHR), adjusted for parental education, smoking and IBD were estimated and pooled using a random-effects model. Antibiotic analyses were adjusted for infection frequency. RESULTS There were 103,046 children (11,872 ABIS and 91,174 MoBa), contributing to 1,663,898 person-years of follow-up, during which 395 were diagnosed with IBD. The frequency of any infection at 0 to < 1 and 1 to < 3 years showed a pooled aHR of 1.01 (95% confidence interval [CI] = 0.96-1.07) and 1.00 (95% CI = 0.99-1.01) per additional infection for IBD. Adjusting for infections, any versus no antibiotics in the first year was associated with IBD (pooled aHR = 1.33 [95% CI = 1.01-1.76]). The aHR for additional antibiotic course was 1.17 (95% CI = 0.96-1.44), driven by penicillin (per additional course, aHR = 1.28 [95% CI = 1.02-1.60]). Although antibiotics at 1 to < 3 years did not show an association with IBD or Crohn's disease, non-penicillin antibiotics were associated with ulcerative colitis (per additional course, aHR = 1.95 [95% CI = 1.38-2.75]). CONCLUSION Early-life antibiotic use was, a significant risk factor for childhood and early adult-onset IBD, independent of infection frequency.
Collapse
Affiliation(s)
- Karl Mårild
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Pediatric Gastroenterology Unit, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tereza Lerchova
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Malin Östensson
- Bioinformatics and Data Centre, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Imberg
- Statistiska Konsultgruppen Sweden, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ketil Størdal
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Children's Center, Oslo University Hospital, Oslo, Norway
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Linköping, Sweden
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
Duan R, Zhang C, Li G, Li J, Duan L. Antibiotic Exposure and Risk of New-Onset Inflammatory Bowel Disease: A Systematic Review and Dose-Response Meta-Analysis. Clin Gastroenterol Hepatol 2025; 23:45-58.e15. [PMID: 38423349 DOI: 10.1016/j.cgh.2024.02.010] [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: 11/10/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND & AIMS The association between antibiotic exposure and inflammatory bowel disease (IBD) remains controversial, especially whether there is a dose-response relationship. We aimed to conduct a systematic review and meta-analysis to thoroughly evaluate the risk of new-onset IBD associated with antibiotic exposure. METHODS Four databases were searched from their inception to September 30, 2023 for all relevant studies. The risk estimates were pooled together using random-effects models, and pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated, stratified by IBD subtype, age, exposure period, study type, and antibiotic classes. Dose-response relationship between the number of antibiotic prescriptions and IBD risk was assessed using generalized least squares regression analysis. RESULTS Twenty-eight studies involving 153,027 patients with IBD were included. Antibiotic exposure was significantly associated with an increased risk of new-onset IBD for prescription-based studies (pooled OR, 1.41; 95% CI, 1.29-1.53) and for questionnaire-based studies (pooled OR, 1.35; 95% CI, 1.08-1.68). This association existed for both Crohn's disease and ulcerative colitis, as well as in children and adults for prescription-based studies. The majority of antibiotic classes were associated with an increased IBD risk, with metronidazole (OR, 1.70; 95% CI, 1.38-2.10) and quinolones (OR, 1.56; 95% CI, 1.37-1.77) having relatively higher risk estimates. A positive nonlinear dose-response association was observed between the number of antibiotic prescriptions and IBD risk. CONCLUSIONS Antibiotic exposure was significantly associated with an increased risk of new-onset IBD, and a positive nonlinear dose-response relationship was observed. Antibiotic stewardship may be important for reducing IBD risk.
Collapse
Affiliation(s)
- Ruqiao Duan
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Haidian District, Beijing, China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Haidian District, Beijing, China
| | - Gaonan Li
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Haidian District, Beijing, China
| | - Jun Li
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Haidian District, Beijing, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Haidian District, Beijing, China.
| |
Collapse
|
6
|
Gu K, Wu A, Liu C, Yu B, He J, Lai X, Chen J, Luo Y, Yan H, Zheng P, Luo J, Pu J, Wang Q, Wang H, Chen D. Absence of gut microbiota alleviates iron overload-induced colitis by modulating ferroptosis in mice. J Adv Res 2024:S2090-1232(24)00608-8. [PMID: 39710300 DOI: 10.1016/j.jare.2024.12.030] [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: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION Iron overload disrupts gut microbiota and induces ferroptosis, contributing to colitis. However, whether gut microbiota directly drives iron overload-induced colitis and its underlying mechanism remain unclear. OBJECTIVES The study aimed to explore whether gut microbiota can directly regulate iron overload-induced colitis and its underling mechanism. METHODS Male C57BL/6N mice were fed with ferrous sulfate to establish an iron overload model. Antibiotics and dextran sulfate sodium salt (DSS) were used to create germ-free and colitis models, respectively. RESULTS Results showed that iron overload caused disruption of systemic iron homeostasis via activating pro-inflammation response, which caused induction of ferroptosis and eventually resulted in colitis in mice. Notably, iron overload inhibited System Xc- and activated the nuclear factor E2-related factor 2/heme oxygenase-1 pathway, driving ferroptosis and colitis progression. Similar results were observed in mouse colon epithelial cells, which were treated with high doses ferric ammonium citrate. Additionally, iron overload exacerbated DSS-induced colitis by activating the ferroptosis and increasing harmful bacteria (e.g., Mucispirillum) abundance. Interestingly, eliminating gut microbiota attenuated iron overload-induced colitis, without affecting systemic inflammation through inhibiting ferroptosis of mice. Depletion of the gut microbiota partially mitigated the exacerbating effect of iron overload on DSS-induced colitis through inhibiting ferroptosis of mice. CONCLUSION Iron overload activates ferroptosis in colonic cells, increases the relative abundance of harmful bacteria, and exacerbates DSS-induced colitis in mice. Iron overload exacerbates DSS-induced ferroptosis and colitis in a microbiota-dependent manner. Targeting gut microbiota may offer new strategies for managing iron overload-induced colitis.
Collapse
Affiliation(s)
- Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Chen Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China; Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junzhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junning Pu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
7
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
8
|
Caron B, Honap S, Peyrin-Biroulet L. Epidemiology of Inflammatory Bowel Disease across the Ages in the Era of Advanced Therapies. J Crohns Colitis 2024; 18:ii3-ii15. [PMID: 39475082 PMCID: PMC11522978 DOI: 10.1093/ecco-jcc/jjae082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND AND AIMS The incidence of inflammatory bowel diseases [IBD] has risen over the past decade to become a global issue. The objectives of this review were to describe the incidence and/or prevalence of IBD in the era of advanced therapies, and to describe the association between environmental risk factors and both pathogenesis and disease course across the ages. METHODS We performed a search of English language publications listed in PubMed regarding the epidemiology of IBD and key environmental factors implicated in IBD from January 2000 to December 2023. RESULTS Annual incidence rates varied by geographical region with IBD estimates ranging from 10.5 to 46.14 per 100 000 in Europe, 1.37 to 1.5 per 100 000 in Asia and the Middle East, 23.67 to 39.8 per 100 000 in Oceania, 0.21 to 3.67 per 100 000 in South America, and 7.3 to 30.2 per 100 000 in North America. The burden of IBD among children and adolescents, and older people is rising globally. Key environmental factors implicated in IBD pathogenesis include exposure to tobacco smoking, antibiotics, non-steroidal anti-inflammatory drugs, oral contraceptives, infections, and ultra-high processed foods. Breastfeeding and a high-quality diet rich in fruit, vegetables, fish, and other fibre sources are important protective factors. Smoking has consistently been shown to negatively impact disease outcomes for Crohn's disease. CONCLUSION The epidemiology of IBD has undergone considerable change in recent decades, with an increase in the burden of disease worldwide. Optimally studying and targeting environmental triggers in IBD may offer future opportunities for disease modification.
Collapse
Affiliation(s)
- Bénédicte Caron
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- INSERM, NGERE, University of Lorraine, F-54000 Nancy, France
- INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
| | - Sailish Honap
- INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- School of Immunology and Microbial Sciences, King’s College London, London, UK
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- INSERM, NGERE, University of Lorraine, F-54000 Nancy, France
- INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Naureckas Li C, Jhaveri R, Scardina T, Patel SJ. Response to "Building the Future of Infectious Diseases: A Call to Action for Quality Improvement Research and Measurement". J Infect Dis 2024; 230:1052-1053. [PMID: 38946348 DOI: 10.1093/infdis/jiae338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024] Open
Affiliation(s)
- Caitlin Naureckas Li
- Division of Infectious Diseases, Ann and Robert H. Lurie Children's Hospital of Chicago
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University
- Center for Quality and Safety
| | - Ravi Jhaveri
- Division of Infectious Diseases, Ann and Robert H. Lurie Children's Hospital of Chicago
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University
| | - Tonya Scardina
- Department of Pharmacy, Ann and Robert H. Lurie Children's Hospital of Chicago, Illinois
| | - Sameer J Patel
- Division of Infectious Diseases, Ann and Robert H. Lurie Children's Hospital of Chicago
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University
- Department of Pharmacy, Ann and Robert H. Lurie Children's Hospital of Chicago, Illinois
| |
Collapse
|
10
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [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] [Indexed: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Ullah H, Ali M, Ma R, Alioui Y, Ali S, Ilyas M, Rahman MU, Ahmed Farooqui N, Siddiqui NZ, Xin Y, Wang L. Polysaccharides derived from Deglet Noor dates modulate amoxicillin-induced dysbiosis and enhance intestinal barrier function. J Funct Foods 2024; 120:106350. [DOI: 10.1016/j.jff.2024.106350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
12
|
Thacker N, Duncanson K, Eslick GD, Dutt S, O'Loughlin EV, Hoedt EC, Collins CE. Antibiotics, passive smoking, high socioeconomic status and sweetened foods contribute to the risk of paediatric inflammatory bowel disease: A systematic review with meta-analysis. J Pediatr Gastroenterol Nutr 2024; 79:610-621. [PMID: 39020449 DOI: 10.1002/jpn3.12303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 07/19/2024]
Abstract
OBJECTIVE Genetic and environmental factors influence pathogenesis and rising incidence of paediatric inflammatory bowel disease (PIBD). The aim was to meta-analyse evidence of diet and environmental factors in PIBD. METHODS A systematic search was conducted to identify diet and environmental factors with comparable risk outcome measures and had been reported in two or more PIBD studies for inclusion in meta-analyses. Those with ≥2 PIBD risk estimates were combined to provide pooled risk estimates. RESULTS Of 4763 studies identified, 36 studies were included. PIBD was associated with higher risk with exposure to ≥/=4 antibiotic courses (includes prescriptions/purchases/courses), passive smoking, not being breastfed, sugary drink intake, being a non-Caucasian child living in a high-income country and infection history (odds ratio [OR] range: 2-3.8). Paediatric Crohn's disease (CD) was associated with higher risk with exposure to antibiotics during early childhood, ≥/=4 antibiotic courses, high socioeconomic status (SES), maternal smoking, history of atopic conditions and infection history (OR range: 1.6-4.4). A history of infection was also associated with higher risk of paediatric ulcerative colitis (UC) (OR: 3.73). Having a higher number of siblings (≥2) was associated with lower risk of paediatric CD (OR: 0.6) and paediatric UC (OR: 0.7). Pet exposure was associated with lower risk of paediatric UC (OR: 0.5). CONCLUSION Several factors associated with PIBD risk were identified that could potentially be used to develop a disease screening tool. Future research is needed to address risk reduction in PIBD.
Collapse
Affiliation(s)
- Nisha Thacker
- School of Health Sciences, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Kerith Duncanson
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
| | - Guy D Eslick
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
| | - Shoma Dutt
- Department of Gastroenterology, The Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, New South Wales, Australia
- Children's Hospital at Westmead Clinical School, Sydney Medical Program, University of Sydney, Sydney, New South Wales, Australia
| | - Edward V O'Loughlin
- Department of Gastroenterology, The Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, New South Wales, Australia
| | - Emily C Hoedt
- NHMRC Centre of Research Excellence in Digestive Health, The University of Newcastle, Sydney, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
| | - Clare E Collins
- School of Health Sciences, College of Health Medicine and Wellbeing, The University of Newcastle, Sydney, New South Wales, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
13
|
Groen J, van der Kuip M, Budding D, Bos MP, Benninga MA, Niemarkt HJ, de Meij TGJ. Assessing Diagnostic Performance of Molecular Culture for Neonatal Sepsis: Protocol of the CHAMPIONS Study. Diagnostics (Basel) 2024; 14:1930. [PMID: 39272715 PMCID: PMC11394283 DOI: 10.3390/diagnostics14171930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Managing neonatal sepsis is challenging due to nonspecific clinical signs, hematological markers with poor accuracy, and a lengthy turnaround time for the identification of microorganisms. Delaying the initiation of antibiotics in truly infected infants can lead to severe morbidity and mortality. Therefore, decisions regarding empiric antibiotic treatment are risk stratified, which exposes many uninfected infants to antibiotics. This causes gut microbiota perturbation, unnecessary hospital admissions, and the generation of multi-resistant organisms. High-speed diagnostic assays could expedite discontinuation or avert the initiation of antibiotics in uninfected infants. This study will evaluate the diagnostic performance of molecular culture (MC), a rapid broad-range PCR-based bacterial profiling technique, for diagnosing neonatal sepsis in infants below 90 days old. A multi-center prospective observational cohort study will include infants evaluated for early and late-onset sepsis. Routine evaluation for suspected sepsis includes microbiological cultures of blood. Additionally, blood for MC will be collected. For early-onset sepsis, umbilical cord blood may be used alternatively. Primary outcome is the agreement between MC and conventional blood culture results. Secondary outcome is the agreement of both assays with clinical sepsis using four different, commonly used definitions. Faster diagnostic pathways for sepsis may reduce antibiotic exposure time. Broad-range molecular assays may identify pathogens undetectable by conventional methods. Employment of umbilical cord blood samples for early-onset sepsis diagnosis can resolve challenges in collecting adequate blood volume and could further expedite treatment decisions.
Collapse
Affiliation(s)
- Jip Groen
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases, Rheumatology and Immunology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | | | | | - Marc A Benninga
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
| | - Hendrik J Niemarkt
- Maxima Medical Center, Department of Neonatology, 5504 DB Veldhoven, The Netherlands
| | - Tim G J de Meij
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
14
|
Jauregui-Amezaga A, Smet A. The Microbiome in Inflammatory Bowel Disease. J Clin Med 2024; 13:4622. [PMID: 39200765 PMCID: PMC11354561 DOI: 10.3390/jcm13164622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
The management of patients with inflammatory bowel disease (IBD) aims to control inflammation through the use of immunosuppressive treatments that target various points in the inflammatory cascade. However, the efficacy of these therapies in the long term is limited, and they often are associated with severe side effects. Although the pathophysiology of the disease is not completely understood, IBD is regarded as a multifactorial disease that occurs due to an inappropriate immune response in genetically susceptible individuals. The gut microbiome is considered one of the main actors in the development of IBD. Gut dysbiosis, characterised by significant changes in the composition and functionality of the gut microbiota, often leads to a reduction in bacterial diversity and anti-inflammatory anaerobic bacteria. At the same time, bacteria with pro-inflammatory potential increase. Although changes in microbiome composition upon biological agent usage have been observed, their role as biomarkers is still unclear. While most studies on IBD focus on the intestinal bacterial population, recent studies have highlighted the importance of other microbial populations, such as viruses and fungi, in gut dysbiosis. In order to modulate the aberrant immune response in patients with IBD, researchers have developed therapies that target different players in the gut microbiome. These innovative approaches hold promise for the future of IBD treatment, although safety concerns are the main limitations, as their effects on humans remain unknown.
Collapse
Affiliation(s)
- Aranzazu Jauregui-Amezaga
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
15
|
Jia Y, Zhang T, He M, Yang B, Wang Z, Liu Y. Melatonin Protects Against Colistin-Induced Intestinal Inflammation and Microbiota Dysbiosis. J Pineal Res 2024; 76:e12989. [PMID: 38978438 DOI: 10.1111/jpi.12989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Colistin is renowned as a last-resort antibiotic due to the emergence of multidrug-resistant pathogens. However, its potential toxicity significantly hampers its clinical utilization. Melatonin, chemically known as N-acetyl-5-hydroxytryptamine, is an endogenous hormone produced by the pineal gland and possesses diverse biological functions. However, the protective role of melatonin in alleviating antibiotic-induced intestinal inflammation remains unknown. Herein, we reveal that colistin stimulation markedly elevates intestinal inflammatory levels and compromises the gut barrier. In contrast, pretreatment with melatonin safeguards mice against intestinal inflammation and mucosal damage. Microbial diversity analysis indicates that melatonin supplementation prevents a reduction in the abundance of Erysipelotrichales and Bifidobacteriales, as well as an increase in Desulfovibrionales abundance, following colistin exposure. Remarkably, short-chain fatty acids (SCFAs) analysis shows that propanoic acid contributes to the protective effect of melatonin on colistin-induced intestinal inflammation. Furthermore, the protection effects of melatonin and propanoic acid on LPS-induced cellular inflammation in RAW 264.7 cells are confirmed. Mechanistic investigations suggest that intervention with melatonin and propanoic acid can repress the activation of the TLR4 signal and its downstream NF-κB and MAPK signaling pathways, thereby mitigating the toxic effects of colistin. Our work highlights the unappreciated role of melatonin in preventing the potential detrimental effects of colistin on intestinal health and suggests a combined therapeutic strategy to effectively manage intestinal infectious diseases.
Collapse
Affiliation(s)
- Yuqian Jia
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tingting Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Mengping He
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Bingqing Yang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
16
|
Mpakosi A, Sokou R, Theodoraki M, Kaliouli-Antonopoulou C. Neonatal Gut Mycobiome: Immunity, Diversity of Fungal Strains, and Individual and Non-Individual Factors. Life (Basel) 2024; 14:902. [PMID: 39063655 PMCID: PMC11278438 DOI: 10.3390/life14070902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
The human gastrointestinal ecosystem, or microbiome (comprising the total bacterial genome in an environment), plays a crucial role in influencing host physiology, immune function, metabolism, and the gut-brain axis. While bacteria, fungi, viruses, and archaea are all present in the gastrointestinal ecosystem, research on the human microbiome has predominantly focused on the bacterial component. The colonization of the human intestine by microbes during the first two years of life significantly impacts subsequent composition and diversity, influencing immune system development and long-term health. Early-life exposure to pathogens is crucial for establishing immunological memory and acquired immunity. Factors such as maternal health habits, delivery mode, and breastfeeding duration contribute to gut dysbiosis. Despite fungi's critical role in health, particularly for vulnerable newborns, research on the gut mycobiome in infants and children remains limited. Understanding early-life factors shaping the gut mycobiome and its interactions with other microbial communities is a significant research challenge. This review explores potential factors influencing the gut mycobiome, microbial kingdom interactions, and their connections to health outcomes from childhood to adulthood. We identify gaps in current knowledge and propose future research directions in this complex field.
Collapse
Affiliation(s)
- Alexandra Mpakosi
- Department of Microbiology, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece
| | - Rozeta Sokou
- Neonatal Intensive Care Unit, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece;
- Neonatal Department, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Martha Theodoraki
- Neonatal Intensive Care Unit, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece;
| | | |
Collapse
|
17
|
Kontou A, Agakidou E, Chatziioannidis I, Chotas W, Thomaidou E, Sarafidis K. Antibiotics, Analgesic Sedatives, and Antiseizure Medications Frequently Used in Critically Ill Neonates: A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:871. [PMID: 39062320 PMCID: PMC11275925 DOI: 10.3390/children11070871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
Antibiotic, analgesic sedative, and antiseizure medications are among the most commonly used medications in preterm/sick neonates, who are at high risk of nosocomial infections, central nervous system complications, and are exposed to numerous painful/stressful procedures. These severe and potentially life-threatening complications may have serious short- and long-term consequences and should be prevented and/or promptly treated. The reported variability in the medications used in neonates indicates the lack of adequate neonatal studies regarding their effectiveness and safety. Important obstacles contributing to inadequate studies in preterm/sick infants include difficulties in obtaining parental consent, physicians' unwillingness to recruit preterm infants, the off-label use of many medications in neonates, and other scientific and ethical concerns. This review is an update on the use of antimicrobials (antifungals), analgesics (sedatives), and antiseizure medications in neonates, focusing on current evidence or knowledge gaps regarding their pharmacokinetics, indications, safety, dosage, and evidence-based guidelines for their optimal use in neonates. We also address the effects of early antibiotic use on the intestinal microbiome and its association with long-term immune-related diseases, obesity, and neurodevelopment (ND). Recommendations for empirical treatment and the emergence of pathogen resistance to antimicrobials and antifungals are also presented. Finally, future perspectives on the prevention, modification, or reversal of antibiotic resistance are discussed.
Collapse
Affiliation(s)
- Angeliki Kontou
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - Eleni Agakidou
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - Ilias Chatziioannidis
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - William Chotas
- Department of Neonatology, University of Vermont, Burlington, VT 05405, USA
| | - Evanthia Thomaidou
- Department of Anesthesia and Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Kosmas Sarafidis
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| |
Collapse
|
18
|
Li Q, Wang J. The Effect of Protein Nutritional Support on Inflammatory Bowel Disease and Its Potential Mechanisms. Nutrients 2024; 16:2302. [PMID: 39064745 PMCID: PMC11280054 DOI: 10.3390/nu16142302] [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: 06/13/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammatory bowel disease (IBD), a complex chronic inflammatory bowel disorder that includes Crohn's disease (CD) and Ulcerative Colitis (UC), has become a globally increasing health concern. Nutrition, as an important factor influencing the occurrence and development of IBD, has attracted more and more attention. As the most important nutrient, protein can not only provide energy and nutrition required by patients, but also help repair damaged intestinal tissue, enhance immunity, and thus alleviate inflammation. Numerous studies have shown that protein nutritional support plays a significant role in the treatment and remission of IBD. This article presents a comprehensive review of the pathogenesis of IBD and analyzes and summarizes the potential mechanisms of protein nutritional support in IBD. Additionally, it provides an overview of the clinical effects of protein nutritional support in IBD and its impact on clinical complications. Research findings reveal that protein nutritional support demonstrates significant benefits in improving clinical symptoms, reducing the risk of complications, and improving quality of life in IBD patients. Therefore, protein nutritional support is expected to provide a new approach for the treatment of IBD.
Collapse
Affiliation(s)
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing 100081, China;
| |
Collapse
|
19
|
Kim TH, Shin JS, Kim SY, Kim J. Association of Previous Antibiotics Use and Kawasaki Disease: A Cohort Study of 106,908 Patients. Pediatr Infect Dis J 2024; 43:643-650. [PMID: 38534913 DOI: 10.1097/inf.0000000000004335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND Microbial imbalance in the gut from antibiotic use may be an etiologic factor of Kawasaki disease (KD). We aimed to identify the association between the use of antibiotics and the development of KD, considering various antibiotic profiles. METHODS A population-based, case-control study was performed using data from the Health Insurance Review and Assessment Service database. Children <5 years of age, who were diagnosed with KD between 2016 and 2019, were identified. Propensity score-matched controls were selected from the general population in a 1:5 ratio. Four separate study cohorts were created according to different periods of antibiotic use: (1) within 28 days and (2) 12 months after birth and (3) within 6 months and (4) 12 months from the index date. Profiles regarding antibiotic use were compared between patients with KD and matched controls. RESULTS We included 17,818 patients with KD and 89,090 matched controls. Use of antibiotics within 6 months [odds ratio (OR): 1.18; 95% confidence interval (CI): 1.12-1.26] and 12 months (OR: 1.23; 95% CI: 1.14-1.32) from the index date were associated with the development of KD. The association between antibiotic use and KD was most prominent in patients who had received 3 or more types of antibiotics within 12 months from the index date (OR: 1.26; 95% CI: 1.17-1.37). CONCLUSIONS Antibiotic use within the preceding 6 or 12 months was associated with KD. Alteration in gut microbiota due to antibiotic usage might play a role in the development of KD.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- From the Spine Center, Department of Orthopedics, Hallym University Sacred Heart Hospital
| | - Ji Seong Shin
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sin Young Kim
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jihye Kim
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Alhindi MY, Almalki FG, Al Saif S, Almalahi A, Alsaegh MH, Mustafa A, AlQurashi MA. Evaluating a Modified Use of the Kaiser Permanente Early-onset Sepsis Risk Calculator to Reduce Antibiotic Exposure: a Retrospective Study. BMJ Paediatr Open 2024; 8:e002597. [PMID: 38844386 PMCID: PMC11163676 DOI: 10.1136/bmjpo-2024-002597] [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: 02/26/2024] [Accepted: 05/12/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Early-onset neonatal sepsis (EONS) remains an important disease entity due to very serious adverse outcomes if left untreated. Lack of diagnostic tools in identifying healthy from diseased neonates, and clinicians' fear of the missing positive-culture sepsis babies, or babies with clinical sepsis have led to overtreating and unnecessary antibiotic exposure. Kaiser Permanente EONS risk calculator is an internally validated tool that can predict EONS. This sepsis risk calculator (SRC) classifies neonates into three subgroups: (1) ill-appearing, (2) equivocal and (3) well-appearing. We propose a modification to this tool that aims to use it solely for well-appearing babies. This modification represents a more conservative approach to decrease antibiotic exposure and offers an alternative for those hesitant to fully implement this tool. METHODS This is a dual-centre retrospective study where data were extracted from the electronic medical records. Our primary outcome was to validate the modified use of the SRC with a two-by-two table. Specificity, negative predictive value and expected antibiotic reduction were used to evaluate the tool's feasibility. RESULT Among 770 babies suspected of EONS, the feasibility of the modified use was tested. The expected antibiotic exposure reduction rate on the modification was 40.4% overall. The proposed modification resulted in a specificity and negative predictive value of 99.28% (95% CI: 97.92% to 99.85%) and 99.5% (95% CI: 99% to 99.8%), respectively. CONCLUSION The modified use of the sepsis risk calculator has shown that it can safely reduce antibiotic exposure in well-appearing babies. The modified use is used as a 'rule out' test that can identify very low risk of EONS babies, and safely minimise antibiotic exposure. Further prospective studies are needed to examine the efficacy of this use, and quality improvement projects are required to evaluate its applicability in different clinical settings.
Collapse
Affiliation(s)
- Mohammed Yasir Alhindi
- Neonatology Division, Department of Pediatrics, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Jeddah, Saudi Arabia
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Faisal Ghazi Almalki
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
| | - Saif Al Saif
- Neonatal Intensive Care Department, Women's Health Specialized Hospital, Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Abdulaziz Almalahi
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Mawaddah Hesham Alsaegh
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
| | - Ahmed Mustafa
- Neonatology Division, Department of Pediatrics, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
| | - Mansour Abdullah AlQurashi
- Neonatology Division, Department of Pediatrics, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Jeddah, Saudi Arabia
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| |
Collapse
|
21
|
Andersen S, Hestetun SV, Bernklev T, Perminow G, Størdal K. Fetal and Early-Life Antibiotics and Risk of Pediatric Inflammatory Bowel Disease: A Population-Based Nationwide Register Study. JOURNAL OF PEDIATRICS. CLINICAL PRACTICE 2024; 12:200096. [PMID: 39949419 PMCID: PMC11824603 DOI: 10.1016/j.jpedcp.2024.200096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2025]
Abstract
Objective To assess whether antibiotic exposure prenatally and before 2 years of age is associated with subsequent pediatric inflammatory bowel disease (PIBD), as earlier studies diverge substantially in risk estimations or do not include prenatal exposure. Methods We performed a register-based cohort study including all children in Norway born 2004-2012 until study's end on December 31, 2020. Exposures were defined as dispensed antibiotics to mothers during pregnancy and to children before 2 years of age. Main outcome was International Classification of Diseases, Tenth Revision, code registrations consistent with PIBD. Results Among 536 819 children, 797 PIBD cases were identified. The aOR for developing PIBD if exposed to antibiotics before 2 years of age compared with no exposure was 1.33 (95% CI 1.15-1.53), adjusted for sex and prenatal antibiotic exposure. An adjustment for maternal smoking during pregnancy increased the aOR to 1.42 (1.22-1.66), but with minimal changes after further adjustments for potential confounders related to pregnancy, birth, or socioeconomic status. A dose-response association was observed in those receiving more than 2 antibiotic courses (aOR 1.47, 1.25-1.73), and with greater effect estimates for broad-spectrum antibiotics (aOR 2.57, 1.82-3.63). Antibiotic exposure during pregnancy was numerically but not significantly associated with offspring PIBD (aOR 1.15, 0.99-1.34). Conclusions Children exposed to antibiotics before 2 years of age were more likely to develop pediatric IBD than controls. Exposure prenatally to maternal antibiotics was numerically but not statistically significant associated to subsequent PIBD.
Collapse
Affiliation(s)
- Svend Andersen
- Department of Pediatrics, Vestfold Hospital Trust, Tønsberg, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sigrid Valen Hestetun
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Tomm Bernklev
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research and Innovation Department, Vestfold Hospital Trust, Tønsberg, Norway
| | - Gøri Perminow
- Pediatric Department, Oslo University Hospital, Oslo, Norway
| | - Ketil Størdal
- Pediatric Department, Oslo University Hospital, Oslo, Norway
- Pediatric Research Institute, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Ananthakrishnan AN, Gerasimidis K, Ho SM, Mayer E, Pollock J, Soni S, Wu GD, Benyacoub J, Ali B, Favreau A, Smith DE, Oh JE, Heller C, Hurtado-Lorenzo A, Moss A, Croitoru K. Challenges in IBD Research 2024: Environmental Triggers. Inflamm Bowel Dis 2024; 30:S19-S29. [PMID: 38778624 DOI: 10.1093/ibd/izae085] [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: 03/15/2024] [Indexed: 05/25/2024]
Abstract
Environmental factors play an important role in inflammatory bowel diseases (IBD; Crohn's disease, [CD], ulcerative colitis [UC]). As part of the Crohn's & Colitis Challenges 2024 agenda, the Environmental Triggers workgroup summarized the progress made in the field of environmental impact on IBD since the last Challenges cycle in this document. The workgroup identified 4 unmet gaps in this content area pertaining to 4 broad categories: (1) Epidemiology; (2) Exposomics and environmental measurement; (3) Biologic mechanisms; and (4) Interventions and Implementation. Within epidemiology, the biggest unmet gaps were in the study of environmental factors in understudied populations including racial and ethnic minority groups and in populations witnessing rapid rise in disease incidence globally. The workgroup also identified a lack of robust knowledge of how environmental factors may impact difference stages of the disease and for different disease-related end points. Leveraging existing cohorts and targeted new prospective studies were felt to be an important need for the field. The workgroup identified the limitations of traditional questionnaire-based assessment of environmental exposure and placed high priority on the identification of measurable biomarkers that can quantify cross-sectional and longitudinal environmental exposure. This would, in turn, allow for identifying the biologic mechanisms of influence of environmental factors on IBD and understand the heterogeneity in effect of such influences. Finally, the working group emphasized the importance of generating high-quality data on effective environmental modification on an individual and societal level, and the importance of scalable and sustainable methods to deliver such changes.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kostantinos Gerasimidis
- Human Nutrition, School of Medicine, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, G31 2ER, Glasgow, UK
| | - Shuk-Mei Ho
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience; Goodman-Luskin Microbiome Center; The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jennifer Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shefali Soni
- Crohn's Disease Program, The Leona M. and Harry B. Helmsley Charitable Trust, New York, NY, USA
| | - Gary D Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Basmah Ali
- Crohn's & Colitis Foundation, IBD Patient Representative, USA
| | - Alex Favreau
- Crohn's & Colitis Foundation, IBD Patient Representative, USA
| | | | - Ji-Eun Oh
- Research Department, Crohn's & Colitis Foundation, New York, NY, USA
| | - Caren Heller
- Research Department, Crohn's & Colitis Foundation, New York, NY, USA
| | | | - Alan Moss
- Research Department, Crohn's & Colitis Foundation, New York, NY, USA
| | - Ken Croitoru
- Division of Gastroenterology, University of Toronto, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
23
|
Li Y, Li Q, Yuan R, Wang Y, Guo C, Wang L. Bifidobacterium breve-derived indole-3-lactic acid ameliorates colitis-associated tumorigenesis by directing the differentiation of immature colonic macrophages. Theranostics 2024; 14:2719-2735. [PMID: 38773969 PMCID: PMC11103503 DOI: 10.7150/thno.92350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Aim: To elucidate dynamics and functions in colonic macrophage subsets, and their regulation by Bifidobacterium breve (B. breve) and its associated metabolites in the initiation of colitis-associated colorectal cancer (CAC). Methods: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to create a CAC model. The tumor-suppressive effect of B. breve and variations of macrophage subsets were evaluated. Intestinal macrophages were ablated to determine their role in the protective effects of B. breve. Efficacious molecules produced by B. breve were identified by non-targeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The molecular mechanism was further verified in murine bone marrow-derived macrophages (BMDMs), macrophages derived from human peripheral blood mononuclear cells (hPBMCs), and demonstrated in CAC mice. Results: B. breve alleviated colitis symptoms, delayed colonic tumorigenesis, and promoted phenotypic differentiation of immature inflammatory macrophages into mature homeostatic macrophages. On the contrary, the ablation of intestinal macrophages largely annulled the protective effects of B. breve. Microbial analysis of colonic contents revealed the enrichment of probiotics and the depletion of potential pathogens following B. breve supplementation. Moreover, indole-3-lactic acid (ILA) was positively correlated with B. breve in CAC mice and highly enriched in the culture supernatant of B. breve. Also, the addition of ILA directly promoted AKT phosphorylation and restricted the pro-inflammatory response of murine BMDMs and macrophages derived from hPBMCs in vitro. The effects of ILA in murine BMDMs and macrophages derived from hPBMCs were abolished by the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the AKT inhibitor MK-2206. Furthermore, ILA could protect against tumorigenesis by regulating macrophage differentiation in CAC mice; the AhR antagonist largely abrogated the effects of B. breve and ILA in relieving colitis and tumorigenesis. Conclusion: B. breve-mediated tryptophan metabolism ameliorates the precancerous inflammatory intestinal milieu to inhibit tumorigenesis by directing the differentiation of immature colonic macrophages.
Collapse
Affiliation(s)
| | | | | | | | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
24
|
Ananthakrishnan AN. Inflammatory bowel diseases: are we ready to recommend a preventive diet for infants? Gut 2024; 73:559-560. [PMID: 38290831 DOI: 10.1136/gutjnl-2023-331849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Simon DA, Kellermayer R. INVITED COMMENTARY on Andersen S, et al. Developmental Windows of Environmental Vulnerability for Inflammatory Bowel Disease. JOURNAL OF PEDIATRICS. CLINICAL PRACTICE 2024; 11:200104. [PMID: 38827481 PMCID: PMC11138252 DOI: 10.1016/j.jpedcp.2024.200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 06/04/2024]
Affiliation(s)
- David A. Simon
- Division of Pediatric Gastroenterology, Texas Children’s Hospital Baylor College of Medicine, Houston, TX
| | - Richard Kellermayer
- Division of Pediatric Gastroenterology, Texas Children’s Hospital Baylor College of Medicine, Houston, TX
- USDA/ARS Children’s Nutrition Research Center, Houston, TX
| |
Collapse
|
26
|
Huang H, Jiang J, Wang X, Jiang K, Cao H. Exposure to prescribed medication in early life and impacts on gut microbiota and disease development. EClinicalMedicine 2024; 68:102428. [PMID: 38312240 PMCID: PMC10835216 DOI: 10.1016/j.eclinm.2024.102428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024] Open
Abstract
The gut microbiota during early life plays a crucial role in infant development. This microbial-host interaction is also essential for metabolism, immunity, and overall human health in later life. Early-life pharmaceutical exposure, mainly referring to exposure during pregnancy, childbirth, and infancy, may change the structure and function of gut microbiota and affect later human health. In this Review, we describe how healthy gut microbiota is established in early life. We summarise the commonly prescribed medications during early life, including antibiotics, acid suppressant medications and other medications such as antidepressants, analgesics and steroid hormones, and discuss how these medication-induced changes in gut microbiota are involved in the pathological process of diseases, including infections, inflammatory bowel disease, metabolic diseases, allergic diseases and neurodevelopmental disorders. Finally, we review some critical methods such as dietary therapy, probiotics, prebiotics, faecal microbiota transplantation, genetically engineered phages, and vagus nerve stimulation in early life, aiming to provide a new strategy for the prevention of adverse health outcomes caused by prescribed medications exposure in early life.
Collapse
Affiliation(s)
- Huan Huang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- Department of Gastroenterology, the Affiliated Jinyang Hospital of Guizhou Medical University, the Second People's Hospital of Guiyang, Guiyang, China
| | - Jiayin Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xinyu Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
27
|
Scholle O, Rasmussen L, Reilev M, Viebrock J, Haug U. Comparative Analysis of Outpatient Antibiotic Prescribing in Early Life: A Population-Based Study Across Birth Cohorts in Denmark and Germany. Infect Dis Ther 2024; 13:299-312. [PMID: 38261237 PMCID: PMC10904695 DOI: 10.1007/s40121-024-00916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
INTRODUCTION Comparing antibiotic prescribing between countries can provide important insights into potential needs of improving antibiotic stewardship programs. We aimed to compare outpatient antibiotic prescribing in early life between children born in Denmark and Germany. METHODS Using the Danish nationwide healthcare registries and a German claims database (GePaRD, ~ 20% population coverage), we included children born between 2004 and 2016, and followed them regarding outpatient antibiotic prescriptions until end of enrollment or the end of 2018. We then determined the median time to first antibiotic prescription. Based on all prescriptions in the first 2 years of life, we calculated the rate of antibiotic treatment episodes and for the children's first prescriptions in this period, we determined established quality indicators. All analyses were stratified by birth year and country. RESULTS In the 2016 birth cohorts, the median time to first antibiotic prescription was ~ 21 months in Denmark and ~ 28 in Germany; the rate of antibiotic treatment episodes per 1000 person-years was 537 in Denmark and 433 in Germany; the percentage of prescribed antibiotics with higher concerns regarding side effects and/or resistance potential was 6.2% in Denmark and 44.2% in Germany. In the 2016 birth cohorts, the age at first antibiotic prescription was 50-59% higher compared to the 2004 birth cohorts; the rate of antibiotic treatment episodes was 43-44% lower. CONCLUSIONS Infants in Denmark received antibiotics markedly earlier and more frequently than in Germany, while quality indicators of antibiotic prescribing were more favorable in Denmark. Although both countries experienced positive changes towards more rational antibiotic prescribing in early life, our findings suggest potential for further improvement. This particularly applies to prescribing antibiotics with a lower potential for side effects and/or resistance in Germany.
Collapse
Affiliation(s)
- Oliver Scholle
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Achterstrasse 30, 28359, Bremen, Germany
| | - Lotte Rasmussen
- Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mette Reilev
- Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jost Viebrock
- Department of Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Ulrike Haug
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Achterstrasse 30, 28359, Bremen, Germany.
- Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany.
| |
Collapse
|
28
|
Jenkins TC, Keith A, Stein AB, Hersh AL, Narayan R, Eggleston A, Rinehart DJ, Patel PK, Walter E, Hargraves IG, Frost HM. Interventions to de-implement unnecessary antibiotic prescribing for ear infections (DISAPEAR Trial): protocol for a cluster-randomized trial. BMC Infect Dis 2024; 24:126. [PMID: 38267837 PMCID: PMC10807124 DOI: 10.1186/s12879-023-08960-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: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Watchful waiting management for acute otitis media (AOM), where an antibiotic is used only if the child's symptoms worsen or do not improve over the subsequent 2-3 days, is an effective approach to reduce antibiotic exposure for children with AOM. However, studies to compare the effectiveness of interventions to promote watchful waiting are lacking. The objective of this study is to compare the effectiveness and implementation outcomes of two pragmatic, patient-centered interventions designed to facilitate use of watchful waiting in clinical practice. METHODS This will be a cluster-randomized trial utilizing a hybrid implementation-effectiveness design. Thirty-three primary care or urgent care clinics will be randomized to one of two interventions: a health systems-level intervention alone or a health systems-level intervention combined with use of a shared decision-making aid. The health systems-level intervention will include engagement of a clinician champion at each clinic, changes to electronic health record antibiotic orders to facilitate delayed antibiotic prescriptions as part of a watchful waiting strategy, quarterly feedback reports detailing clinicians' use of watchful waiting individually and compared with peers, and virtual learning sessions for clinicians. The hybrid intervention will include the health systems-level intervention plus a shared decision-making aid designed to inform decision-making between parents and clinicians with best available evidence. The primary outcomes will be whether an antibiotic was ultimately taken by the child and parent satisfaction with their child's care. We will explore the differences in implementation effectiveness by patient population served, clinic type, clinical setting, and organization. The fidelity, acceptability, and perceived appropriateness of the interventions among different clinician types, patient populations, and clinical settings will be compared. We will also conduct formative qualitative interviews and surveys with clinicians and administrators, focus groups and surveys of parents of patients with AOM, and engagement of two stakeholder advisory councils to further inform the interventions. DISCUSSION This study will compare the effectiveness of two pragmatic interventions to promote use of watchful waiting for children with AOM to reduce antibiotic exposure and increase parent satisfaction, thus informing national antibiotic stewardship policy development. CLINICAL TRIAL REGISTRATION NCT06034080.
Collapse
Affiliation(s)
- Timothy C Jenkins
- Division of Infectious Diseases, Department of Medicine, Denver Health and Hospital Authority, Denver, CO, USA
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Amy Keith
- Center for Health Systems Research, Denver Health and Hospital Authority, 601 Broadway Ave, Denver, CO, USA.
| | - Amy B Stein
- Center for Health Systems Research, Denver Health and Hospital Authority, 601 Broadway Ave, Denver, CO, USA
| | - Adam L Hersh
- Division of Infectious Diseases, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | | | | | - Deborah J Rinehart
- Center for Health Systems Research, Denver Health and Hospital Authority, 601 Broadway Ave, Denver, CO, USA
- Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Payal K Patel
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Health, Murray, UT, USA
| | | | - Ian G Hargraves
- Knowledge and Evaluation Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Holly M Frost
- Center for Health Systems Research, Denver Health and Hospital Authority, 601 Broadway Ave, Denver, CO, USA.
- Department of Pediatrics, Denver Health and Hospital Authority, 601 Broadway Ave, Denver, CO, USA.
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
29
|
Bradley E, Haran J. The human gut microbiome and aging. Gut Microbes 2024; 16:2359677. [PMID: 38831607 PMCID: PMC11152108 DOI: 10.1080/19490976.2024.2359677] [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: 01/19/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The composition of the human gut microbiome has been observed to change over the course of an individual's life. From birth, it is shaped by mode of delivery, diet, environmental exposures, geographic location, exposures to medications, and by aging itself. Here, we present a narrative review of the gut microbiome across the lifespan with a focus on its impacts on aging and age-related diseases in humans. We will describe how it is shaped, and features of the gut microbiome that have been associated with diseases at different phases of life and how this can adversely affect healthy aging. Across the lifespan, and especially in old age, a diverse microbiome that includes organisms suspected to produce anti-inflammatory metabolites such as short-chain fatty acids, has been reported to be associated with healthy aging. These findings have been remarkably consistent across geographic regions of the world suggesting that they could be universal features of healthy aging across all cultures and genetic backgrounds. Exactly how these features of the microbiome affect biologic processes associated with aging thus promoting healthy aging will be crucial to targeting the gut microbiome for interventions that will support health and longevity.
Collapse
Affiliation(s)
- Evan Bradley
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| | - John Haran
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| |
Collapse
|
30
|
Krueger C, Alqurashi W, Barrowman N, Litwinska M, Le Saux N. The long and the short of pediatric emergency department antibiotic prescribing: A retrospective observational study. Am J Emerg Med 2024; 75:131-136. [PMID: 37950980 DOI: 10.1016/j.ajem.2023.10.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND Most antibiotics prescribed to children are provided in the outpatient and emergency department (ED) settings, yet these prescribers are seldom engaged by antibiotic stewardship programs. We reviewed ED antibiotic prescriptions for three common infections to describe current prescribing practices. METHODS Prescription data between 2018 and 2021 were extracted from the electronic records of children discharged from the Children's Hospital of Eastern Ontario ED with urinary tract infection (UTI), community acquired pneumonia (CAP), and acute otitis media ≥2 years of age (AOM). Antibiotic choice, duration, as well as the provider's time in practice and training background were collected. Antibiotic durations were compared with Canadian guideline recommendations to assess concordance. Provider-level prescribing practices were analyzed using k-means cluster analysis. RESULTS 10,609 prescriptions were included: 2868 for UTI, 2958 for CAP, and 4783 for AOM. Guideline-concordant durations prescribed was generally high (UTI 84.9%, CAP 94.0%, AOM 52.8%), a large proportion of antibiotic-days prescribed were in excess of the minimally recommended duration for each infection (UTI 16.8%, 19.3%, AOM 25.5%). Cluster analysis yielded two clusters of prescribers, with those in one cluster more commonly prescribing durations at the lower end of recommended interval, and the others more commonly prescribing longer durations for all three infections reviewed. No statistically significant differences were found between clusters by career stage or training background. CONCLUSIONS While guideline-concordant antibiotic prescribing was generally high, auditing antibiotic prescriptions identified shifting prescribing towards the minimally recommended duration as a potential opportunity to reduce antibiotic use among children for these infections.
Collapse
Affiliation(s)
- Carsten Krueger
- Division of Infectious Diseases, Immunology & Allergy, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.
| | - Waleed Alqurashi
- Division of Emergency Medicine, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Nicholas Barrowman
- Children's Hospital of Eastern Ontario Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Maria Litwinska
- Business Intelligence Team, Information Services, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Nicole Le Saux
- Division of Infectious Diseases, Immunology & Allergy, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
31
|
Coutry N, Gasmi I, Herbert F, Jay P. Mechanisms of intestinal dysbiosis: new insights into tuft cell functions. Gut Microbes 2024; 16:2379624. [PMID: 39042424 PMCID: PMC11268228 DOI: 10.1080/19490976.2024.2379624] [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: 12/18/2023] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Symbiosis between the host and intestinal microbial communities is essential for human health. Disruption in this symbiosis is linked to gastrointestinal diseases, including inflammatory bowel diseases, as well as extra-gastrointestinal diseases. Unbalanced gut microbiome or gut dysbiosis contributes in multiple ways to disease frequency, severity and progression. Microbiome taxonomic profiling and metabolomics approaches greatly improved our understanding of gut dysbiosis features; however, the precise mechanisms involved in gut dysbiosis establishment still need to be clarified. The aim of this review is to present new actors and mechanisms underlying gut dysbiosis formation following parasitic infection or in a context of altered Paneth cells, revealing the existence of a critical crosstalk between Paneth and tuft cells to control microbiome composition.
Collapse
Affiliation(s)
- Nathalie Coutry
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Imène Gasmi
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Fabien Herbert
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Philippe Jay
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| |
Collapse
|
32
|
Kim N. Colorectal Diseases and Gut Microbiome. SEX/GENDER-SPECIFIC MEDICINE IN CLINICAL AREAS 2024:137-208. [DOI: 10.1007/978-981-97-0130-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Khalessi A, Crowe BR, Xia Y, Rubinfeld G, Baylor J, Radin A, Liang PS, Chen LA. Differential Manifestations of Inflammatory Bowel Disease Based on Race and Immigration Status. GASTRO HEP ADVANCES 2023; 3:326-332. [PMID: 38765199 PMCID: PMC11101196 DOI: 10.1016/j.gastha.2023.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 11/26/2023] [Indexed: 05/21/2024]
Abstract
BACKGROUND AND AIMS The prevalence of inflammatory bowel disease (IBD) is increasing globally. In this context, identifying risk factors for severe disease is important. We examined how race/ethnicity and immigration status influence IBD manifestations, treatments, and outcomes in a diverse, tertiary-care safety-net hospital. METHODS We conducted a single-center retrospective review of all IBD inpatients and outpatients treated from 1997-2017. Using logistic regression modeling, we compared disease onset, treatment, and outcomes by race (White, Black, Hispanic, or Asian) and immigration status (US-born vs foreign-born). RESULTS A total of 577 patients were identified, of which 29.8% were White, 27.4% were Hispanic, 21.7% were Black, and 13.0% were Asian. Compared to Whites, Asians were more likely to be male (odds ratio [OR] 2.63, 95% confidence interval [CI]: 1.45, 5.00), whereas Blacks were more likely to be diagnosed with Crohn's disease (OR 1.75, 95% CI: 1.10, 2.77) and more likely to undergo IBD-related intestinal resection (OR 2.49, 95% CI: 1.40, 4.50). Compared to US-born patients, foreign-born patients were more likely to be diagnosed with ulcerative colitis (OR 1.77, 95% CI: 1.04, 3.02). They were also less likely to be diagnosed before 16 years of age (OR 0.19, 95% CI: 0.08, 0.41), to have undergone intestinal resections (OR 0.39, 95% CI: 0.19, 0.83), to have received biologics (OR 0.43, 95% CI: 0.25, 0.76), or to have had dermatologic manifestations (OR 0.12, 95% CI: 0.03, 0.41). CONCLUSION IBD phenotype varies by race, although foreign-born patients of all races show evidence of later-onset and milder disease. These findings may aid in disease prognostication and clinical management and, furthermore, may provide insight into intrinsic and environmental influences on IBD pathogenesis.
Collapse
Affiliation(s)
- Ali Khalessi
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Brooks R. Crowe
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Yuhe Xia
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, New York
| | - Gregory Rubinfeld
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Jessica Baylor
- Division of Gastroenterology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Arielle Radin
- Division of Gastroenterology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Peter S. Liang
- Division of Gastroenterology, Department of Medicine, New York University School of Medicine, New York, New York
- VA New York Harbor Health Care System, New York, New York
| | - Lea Ann Chen
- Division of Gastroenterology, Department of Medicine, New York University School of Medicine, New York, New York
- NYC Health + Hospitals/Bellevue, New York, New York
| |
Collapse
|
34
|
Bertin B, Foligne B, Ley D, Lesage J, Beghin L, Morcel J, Gottrand F, Hermann E. An Overview of the Influence of Breastfeeding on the Development of Inflammatory Bowel Disease. Nutrients 2023; 15:5103. [PMID: 38140362 PMCID: PMC10745409 DOI: 10.3390/nu15245103] [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: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The first 1000 days of life is a critical period that contributes significantly to the programming of an individual's future health. Among the many changes that occur during this period early in life, there is growing evidence that the establishment of healthy gut microbiota plays an important role in the prevention of both short- and long-term health problems. Numerous publications suggest that the quality of the gut microbiota colonisation depends on several dietary factors, including breastfeeding. In this respect, a relationship between breastfeeding and the risk of inflammatory bowel disease (IBD) has been suggested. IBDs are chronic intestinal diseases, and perinatal factors may be partly responsible for their onset. We review the existence of links between breastfeeding and IBD based on experimental and clinical studies. Overall, despite encouraging experimental data in rodents, the association between breastfeeding and the development of IBD remains controversial in humans, partly due to the considerable heterogeneity between clinical studies. The duration of exclusive breastfeeding is probably decisive for its lasting effect on IBD. Thus, specific improvements in our knowledge could support dietary interventions targeting the gut microbiome, such as the early use of prebiotics, probiotics or postbiotics, in order to prevent the disease.
Collapse
Affiliation(s)
- Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Benoit Foligne
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| |
Collapse
|
35
|
Abstract
Most antibiotics are prescribed in ambulatory setting and at least 30% to 50% of these prescriptions are unnecessary. The use of antibiotics when not needed promotes the development of antibiotic resistant organisms and harms patients by placing them at risk for adverse drug events and Clostridioides difficile infections. National guidelines recommend that health systems implement antibiotic stewardship programs in ambulatory settings. However, uptake of stewardship in ambulatory setting has remained low. This review discusses the current state of ambulatory stewardship in the United States, best practices for the successful implementation of effective ambulatory stewardship programs, and future directions to improve antibiotic use in ambulatory settings.
Collapse
Affiliation(s)
- Holly M Frost
- Center for Health Systems Research, Denver Health and Hospital Authority, Denver, CO, USA; Department of Pediatrics, Denver Health and Hospital Authority, Denver, CO, USA; Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Adam L Hersh
- Division of Infectious Disease, Department of Pediatrics, 295 Chipeta Way, Salt Lake City, UT 8413, USA
| | - David Y Hyun
- Antimicrobial Resistance Project, The Pew Charitable Trusts, 901 East Street NW, Washington, DC 20004-2008, USA
| |
Collapse
|
36
|
Chowdhury F, Hill L, Shah N, Popov J, Cheveldayoff P, Pai N. Intestinal microbiome in short bowel syndrome: diagnostic and therapeutic opportunities. Curr Opin Gastroenterol 2023; 39:463-471. [PMID: 37751391 DOI: 10.1097/mog.0000000000000970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW The intestinal microbiome plays a strong, complementary role in the development and integrity of the intestinal epithelium. This biology is crucial for intestinal adaptation, particularly after the mucosal insults that lead to short bowel syndrome (SBS). The purpose of this review is to discuss relationships between the intestinal microbiota and the physiology of intestinal adaptation. RECENT FINDINGS We will address interactions between the intestinal microbiome and nutritional metabolism, factors leading to dysbiosis in SBS, and common compositional differences of the gut microbiome in SBS patients as compared to healthy controls. We will also discuss novel opportunities to expand diagnostic and therapeutic interventions in this population, by using our knowledge of the microbiome to manipulate luminal bacteria and study their resultant metabolites. As microbial therapeutics advance across so many fields of medicine, this review is timely in its advocacy for ongoing research that focuses on the SBS population.Our review will discuss 4 key areas: 1) physiology of the intestinal microbiome in SBS, 2) clinical and therapeutic insults that lead to a state of dysbiosis, 3) currently available evidence on microbiome-based approaches to SBS management, and 4) opportunities and innovations to inspire future research. SUMMARY The clinical implications of this review are both current, and potential. Understanding how the microbiome impacts intestinal adaptation and host physiology may enhance our understanding of why we experience such clinical variability in SBS patients' outcomes. This review may also expand clinicians' understanding of what 'personalized medicine' can mean for this patient population, and how we may someday consider our nutritional, therapeutic, and prognostic recommendations based on our patients' host, and microbial physiology.
Collapse
Affiliation(s)
- Fariha Chowdhury
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
| | - Lee Hill
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Department of Pediatrics, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Nyah Shah
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
| | - Jelena Popov
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paige Cheveldayoff
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Centre for Metabolism, Obesity and Diabetes Research
| | - Nikhil Pai
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Centre for Metabolism, Obesity and Diabetes Research
- Farncombe Family Digestive Health Research Institute, McMaster University
- Division of Pediatric Gastroenterology & Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
37
|
Hong JY, Medzhitov R. On developmental programming of the immune system. Trends Immunol 2023; 44:877-889. [PMID: 37852863 DOI: 10.1016/j.it.2023.09.004] [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: 09/05/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 10/20/2023]
Abstract
Early-life environmental exposures play a significant role in shaping long-lasting immune phenotypes and disease susceptibility. Nevertheless, comprehensive understanding of the developmental programming of immunity is limited. We propose that the vertebrate immune system contains durable programmable components established through early environmental interactions and maintained in a stable and homeostatic manner. Some immune components, such as immunological memory, are intrinsically programmable. Others are influenced by conditions during critical developmental windows in early life, including microbiota, hormones, metabolites, and environmental stress, which impact programming. Developmental immune programming can promote adaptation to an anticipated future environment. However, mismatches between predicted and actual environments can result in disease. This is relevant because understanding programming mechanisms can offer insights into the origin of inflammatory diseases, ideally enabling effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Jun Young Hong
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
38
|
Keith A, Jenkins TC, O'Leary S, Stein AB, Katz SE, Newland J, Rinehart DJ, Gilbert A, Dodd S, Terrill CM, Frost HM. Reducing length of antibiotics for children with ear infections: protocol for a cluster-randomized trial in the USA. J Comp Eff Res 2023; 12:e230088. [PMID: 37855227 PMCID: PMC10690393 DOI: 10.57264/cer-2023-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Aim: Preventing unnecessarily long durations of antibiotic therapy is a key opportunity to reduce antibiotic overuse in children 2 years of age and older with acute otitis media (AOM). Pragmatic interventions to reduce durations of therapy that can be effectively scaled and sustained are urgently needed. This study aims to fill this gap by evaluating the effectiveness and implementation outcomes of two low-cost interventions of differing intensities to increase guideline-concordant antibiotic durations in children with AOM. Methods: The higher intensity intervention will consist of clinician education regarding guideline-recommended short durations of antibiotic therapy; electronic health record (EHR) prescription field changes to promote prescribing of recommended short durations; and individualized clinician audit and feedback on adherence to recommended short durations of therapy in comparison to peers, while the lower intensity intervention will consist only of clinician education and EHR changes. We will explore the differences in implementation effectiveness by patient population served, clinician type, clinical setting and organization as well as intervention type. The fidelity, feasibility, acceptability and perceived appropriateness of the interventions among different clinician types, patient populations, clinical settings and intervention type will be compared. We will also conduct formative qualitative interviews with clinicians and administrators and focus groups with parents of patients to further inform the interventions and study. The formative evaluation will take place over 1.5 years, the interventions will be implemented over 2 years and evaluation of the interventions will take place over 1.5 years. Discussion: The results of this study will provide a framework for other healthcare systems to address the widespread problem of excessive durations of therapy for AOM and inform national antibiotic stewardship policy development. Clinical Trial Registration: NCT05608993 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Amy Keith
- Center for Health Systems Research, Denver Health & Hospital Authority, Denver, CO 80201, USA
| | - Timothy C Jenkins
- Division of Infectious Diseases, Department of Medicine, Denver Health & Hospital Authority, Denver, CO 80204, USA
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Sonja O'Leary
- Department of General Pediatrics, Denver Health Medical Center, Denver, CO 80204, USA
- Department of General Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Amy B Stein
- Center for Health Systems Research, Denver Health & Hospital Authority, Denver, CO 80201, USA
| | - Sophie E Katz
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jason Newland
- Division of Infectious Diseases, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Deborah J Rinehart
- Center for Health Systems Research, Denver Health & Hospital Authority, Denver, CO 80201, USA
- Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045,, USA
| | - Aiden Gilbert
- Center for Health Systems Research, Denver Health & Hospital Authority, Denver, CO 80201, USA
| | - Sherry Dodd
- Division of Infectious Diseases, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Cindy M Terrill
- Division of Infectious Diseases, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Holly M Frost
- Center for Health Systems Research, Denver Health & Hospital Authority, Denver, CO 80201, USA
- Department of General Pediatrics, Denver Health Medical Center, Denver, CO 80204, USA
- Department of General Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
39
|
Khan R, Kuenzig ME, Benchimol EI. Epidemiology of Pediatric Inflammatory Bowel Disease. Gastroenterol Clin North Am 2023; 52:483-496. [PMID: 37543395 DOI: 10.1016/j.gtc.2023.05.001] [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] [Indexed: 08/07/2023]
Abstract
Inflammatory bowel disease (IBD), including subtypes Crohn disease and ulcerative colitis is a chronic inflammatory disorder most often diagnosed in young adulthood. The incidence and prevalence of pediatric-onset IBD is increasing globally. IBD is likely caused by an interplay of multiple environmental factors resulting in a dysregulated mucosal response to the commensal intestinal microbiota in genetically predisposed individuals. This article provides an overview of pediatric IBD epidemiology and environmental risk factors associated with its development, such as the Hygiene Hypothesis, air pollution, greenspace and blue space, neonatal factors, antibiotics, and diet.
Collapse
Affiliation(s)
- Rabia Khan
- SickKids Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children (SickKids), 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - M Ellen Kuenzig
- SickKids Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children (SickKids), 555 University Avenue, Toronto, ON M5G 1X8, Canada; Child Health Evaluative Sciences, SickKids Research Institute
| | - Eric I Benchimol
- SickKids Inflammatory Bowel Disease Centre, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children (SickKids), 555 University Avenue, Toronto, ON M5G 1X8, Canada; Child Health Evaluative Sciences, SickKids Research Institute; ICES, Toronto, Canada; Department of Paediatrics and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada.
| |
Collapse
|
40
|
Narula N, Wong ECL, Pray C, Marshall JK, Rangarajan S, Islam S, Bahonar A, Alhabib KF, Kontsevaya A, Ariffin F, Co HU, Al Sharief W, Szuba A, Wielgosz A, Diaz ML, Yusuf R, Kruger L, Soman B, Li Y, Wang C, Yin L, Mirrakhimov E, Lanas F, Davletov K, Rosengren A, Lopez-Jaramillo P, Khatib R, Oguz A, Iqbal R, Yeates K, Avezum Á, Reinisch W, Moayyedi P, Yusuf S. Associations of Antibiotics, Hormonal Therapies, Oral Contraceptives, and Long-Term NSAIDS With Inflammatory Bowel Disease: Results From the Prospective Urban Rural Epidemiology (PURE) Study. Clin Gastroenterol Hepatol 2023; 21:2649-2659.e16. [PMID: 36528284 DOI: 10.1016/j.cgh.2022.11.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/20/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND & AIMS Several medications have been suspected to contribute to the etiology of inflammatory bowel disease (IBD). This study assessed the association between medication use and the risk of developing IBD using the Prospective Urban Rural Epidemiology cohort. METHODS This was a prospective cohort study of 133,137 individuals between the ages of 20 and 80 from 24 countries. Country-specific validated questionnaires documented baseline and follow-up medication use. Participants were followed up prospectively at least every 3 years. The main outcome was the development of IBD, including Crohn's disease (CD) and ulcerative colitis (UC). Short-term (baseline but not follow-up use) and long-term use (baseline and subsequent follow-up use) were evaluated. Results are presented as adjusted odds ratios (aORs) with 95% CIs. RESULTS During a median follow-up period of 11.0 years (interquartile range, 9.2-12.2 y), there were 571 incident IBD cases (143 CD and 428 UC). Incident IBD was associated significantly with baseline antibiotic (aOR, 2.81; 95% CI, 1.67-4.73; P = .0001) and hormonal medication use (aOR, 4.43; 95% CI, 1.78-11.01; P = .001). Among females, previous or current oral contraceptive use also was associated with IBD development (aOR, 2.17; 95% CI, 1.70-2.77; P < .001). Nonsteroidal anti-inflammatory drug users also were observed to have increased odds of IBD (aOR, 1.80; 95% CI, 1.23-2.64; P = .002), which was driven by long-term use (aOR, 5.58; 95% CI, 2.26-13.80; P < .001). All significant results were consistent in direction for CD and UC with low heterogeneity. CONCLUSIONS Antibiotics, hormonal medications, oral contraceptives, and long-term nonsteroidal anti-inflammatory drug use were associated with increased odds of incident IBD after adjustment for covariates.
Collapse
Affiliation(s)
- Neeraj Narula
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton Health Sciences, Hamilton, Ontario, Canada.
| | - Emily C L Wong
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Cara Pray
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - John K Marshall
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Sumathy Rangarajan
- Population Health Research Institute, McMaster University, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Shofiqul Islam
- Population Health Research Institute, McMaster University, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Ahmad Bahonar
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Khalid F Alhabib
- Department of Cardiac Sciences, King Fahad Cardiac Center, College of Medicine, King Saud Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Anna Kontsevaya
- National Research Center for Therapy and Preventive Medicine, Russian Federation, Moscow, Russia
| | - Farnaza Ariffin
- Primary Care Medicine, Faculty of Medicine Universiti Teknologi MARA, Selangor, Malaysia
| | - Homer U Co
- University of the Philippines College of Medicine, Manila, Philippines
| | - Wadeia Al Sharief
- Family Medicine Department, Medical Education and Research Department, Dubai Health Authority, Dubai, United Arab Emirates
| | - Andrzej Szuba
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Wroclaw, Poland
| | | | - Maria Luz Diaz
- Estudios Clínicos Latinoamérica Rosario, Santa Fe, Argentina
| | - Rita Yusuf
- Independent University, Bashundhara R/A, Dhaka, Bangladesh
| | - Lanthé Kruger
- Africa Unit for Transdisciplinary Health Research, Potchefstroom, South Africa
| | - Biju Soman
- Health Action by People, Thiruvananthapuram, Achutha Menon Centre for Health Science Studies, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Yang Li
- Medical Research and Biometrics Center, National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chuangshi Wang
- Medical Research and Biometrics Center, National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lu Yin
- Medical Research and Biometrics Center, National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Kairat Davletov
- Al-Farabi Kazakh National University, Health Research Institute, Almaty, Kazakhstan
| | - Annika Rosengren
- Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Västra Götaland Region Region, Sweden
| | | | - Rasha Khatib
- Advocate Aurora Research Institute, Advocate Aurora Health, Downers Grove, Illinois; Institute of Community and Public Health, Birzeit University, Birzeit, Palestine
| | - Aytekin Oguz
- Istanbul Medeniyet University, Faculty of Medicine, Department of Internal Medicine, Istanbul, Turkey
| | - Romaina Iqbal
- Department of Community Health Sciences, Aga Khan University, Karachi City, Sindh, Pakistan
| | - Karen Yeates
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Álvaro Avezum
- International Research Center, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
| | - Walter Reinisch
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Paul Moayyedi
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Salim Yusuf
- Population Health Research Institute, McMaster University, Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
41
|
Jawad AB, Jansson S, Wewer V, Malham M. Early Life Oral Antibiotics Are Associated With Pediatric-Onset Inflammatory Bowel Disease-A Nationwide Study. J Pediatr Gastroenterol Nutr 2023; 77:366-372. [PMID: 37346028 DOI: 10.1097/mpg.0000000000003861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
OBJECTIVES Early-life environmental triggers are thought to play a larger role in pediatric-onset inflammatory bowel disease (pIBD) compared to adult-onset IBD. We aimed to assess the risk of developing pIBD after exposure to oral antibiotics during the first 5 years of life. METHODS In a nation-wide cohort study, we identified all patients diagnosed with pIBD (<18 years at diagnosis) in Denmark between 1995 and 2018 from the National Patient Registry and matched them with up to 10 reference individuals. Antibiotic exposure was defined as being prescribed antibiotics during first 5 years of life. Data were retrieved from the National Prescription Register. Outcome was developing pIBD. Risk estimates are presented by hazard ratios (HR) with 95% confidence intervals (CI). RESULTS We identified 1927 pIBD patients and 18,318 reference individuals. Oral antibiotic exposure during the first 5 years of life was associated with a higher risk of developing pIBD (HR = 1.33 [95% CI: 1.2-1.5], P <0.0001). The risk was also increased if patients had ≥4 antibiotic prescriptions compared to no antibiotics (HR = 1.33 [95% CI: 1.2-1.5], P <0.0001). Broad-spectrum antibiotics increased the risk of pIBD compared to narrow-spectrum antibiotics (HR = 1.29 [95% CI: 1.2-1.4], P < 0.0001). When stratified by IBD subtypes, only Crohn disease was significantly associated with exposure to antibiotics (HR = 1.37 [95% CI: 1.1-1.7], P = 0.002). CONCLUSIONS In this nationwide registry-based study, we found that oral antibiotic exposure during first 5 years of life was associated with an increased risk of pIBD. Repeated antibiotic exposures increased risk estimates.
Collapse
Affiliation(s)
- Ali Bashir Jawad
- From the Medical Faculty, University of Copenhagen, Copenhagen, Denmark
- the Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
| | - Sabine Jansson
- the Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
- the Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
| | - Vibeke Wewer
- the Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
- the Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
| | - Mikkel Malham
- the Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
- the Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Hvidovre, Hvidovre, Denmark
- the Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Nath S, Alissa R, Shukla S, Li M, Smotherman C, Hudak ML. Tailored Approach to Evaluation and Management of Early Onset Neonatal Sepsis in a Safety-Net Teaching Hospital in Northeast Florida. Cureus 2023; 15:e45263. [PMID: 37846280 PMCID: PMC10576972 DOI: 10.7759/cureus.45263] [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: 03/30/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
Objective Early onset neonatal sepsis (EONS) remains a significant cause of morbidity and mortality in newborns in the immediate postnatal period. High empiric antibiotic use in well-appearing infants with known risk factors for sepsis led the American Academy of Pediatrics (AAP) to revise its 2010 guidelines for the evaluation and management of EONS to avoid overuse of antibiotics. In this recent clinical report, the AAP provided a framework that outlined several evidence-based approaches for sepsis risk assessment in newborns that can be adopted by institutions based on local resources and structure. One of these approaches, the sepsis risk calculator (SRC) developed by Kaiser Permanente, has been widely validated for reducing unnecessary antibiotic exposure and blood work in infants suspected of having EONS. In order to determine the utility and safety of modifying our institution's protocol to the SRC, we implemented a two-phased approach to evaluate the use of SRC in our newborn nursery. Phase 1 utilized a retrospective review of cases with SRC superimposition. If results from Phase 1 were found to be favorable, Phase 2 initiated a trial of the SRC for a six-month period prior to complete implementation. Methods Phase 1 consisted of retrospectively applying the SRC to electronic medical records (EMR) of infants ≥ 35 weeks' gestational age admitted to the newborn nursery with risk factors for EONS between June 2016 and May 2017. We compared actual antibiotic use as determined by the unit's EONS protocol for evaluation and management based on 2010 Centers for Disease Control and Prevention (CDC) and AAP guidelines to SRC-recommended antibiotic use. We used EMR to determine maternal and infant data, blood work results, and antibiotic usage as well as used daily progress notes by the clinical team to determine the clinical status of the infants retrospectively. Based on the projected reduction in blood work and antibiotics use with the retrospective superimposition of SRC on this cohort of infants and identification of our high-risk patient subset, we developed a novel, hybrid EONS protocol that we implemented and assessed throughout Phase 2, a six-month period from August 2018 to January 2019, as a prospective observational study. Results Phase 1 (SRC superimposition) demonstrated that the use of the SRC would have reduced empiric antibiotic use from 56% to 13% in the study cohort when compared with 2010 CDC/AAP guidelines. However, these same findings revealed use of the SRC would have resulted in delayed evaluation and initiation of antibiotics in 2 of 4 chorioamnionitis-exposed infants with positive blood cultures. During Phase 2 (n=302), with the implementation of our tailored approach (SRC implementation with additional blood culture in chorioamnionitis-exposed infants), 12 (4%) neonates received empiric antibiotic treatment compared to nine (3%) neonates who would have been treated per strict adherence to SRC recommendations. No neonate had culture-positive EONS. Continued use of 2010 CDC/AAP guidelines would have led to empiric antibiotic use in 38 (12.6%) infants in this cohort. Conclusion We developed a novel hybrid approach to the evaluation and management of neonates at increased risk of EONS by tailoring SRC recommendations to our safety-net population. Our stewardship effort achieved a safe and significant reduction in antibiotic usage compared to prior usage determined using CDC/AAP guidelines.
Collapse
Affiliation(s)
- Sfurti Nath
- Pediatrics/Neonatal Perinatal Medicine, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Rana Alissa
- Pediatrics, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | | | - Meng Li
- Pediatrics, Pediatric First, Warner Robins, USA
| | - Carmen Smotherman
- Pathology/Biostatistics, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Mark L Hudak
- Pediatrics/Neonatal Perinatal Medicine, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| |
Collapse
|
43
|
Girma A. The effect of novel antimicrobial agents on the normal functioning of human intestinal microbiota: a systematic review. FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1159352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Antimicrobial agents have significant effects on the ecological balance of the human microbiota through incomplete absorption (e.g., orally administered antimicrobial agents) or secretion (e.g., by the salivary glands, in the bile, or from the intestinal mucosa) of the agents. This study aimed to examine the effects of novel antimicrobial agents on the normal functioning of the intestinal microbiota. The articles, written in English, were recovered from PubMed, ScienceDirect, Web of Science, Google Scholar, and DOAJ, as well as from manual searches using a reference list. “Microbiota”, “Intestinal Microbiota”, “Eubiotic Microbiota”, “Ecological Impact”, “Antimicrobial Agents,”, “Antibiotics”, “Dysbiosis”, “Gut Microbiota”, and “Probiotics” were the search terms used to retrieve the articles. The PRISMA 2009 checklist was applied for article search strategy, article selection, data extraction, and result reporting for the review process. A total of eight original research articles were included from a total of 379 articles obtained in different search strategies. The eight new antimicrobial agents demonstrated significant impacts on the ecological balance of the human intestinal microbiota. Therefore, eubiosis is crucial in preventing the establishment of exogenous antimicrobial-resistant strains as well as their gene transfer.Systematic review registration[PRISMA], identifier [2009].
Collapse
|
44
|
Oh SJ, Kim HJ, Lee CK. A dose-dependent increase in the risk of inflammatory bowel disease after exposure to broad-spectrum antibiotics: A national population study in Korea. Aliment Pharmacol Ther 2023; 58:191-206. [PMID: 37154240 DOI: 10.1111/apt.17542] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/26/2022] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND The association between antibiotic use and risk of inflammatory bowel disease (IBD), particularly among adults, remains unclear. Furthermore, there is a scarcity of data among non-Western countries. AIMS To investigate the association and dose-response relationships between antibiotic use and subsequent IBD risk across all ages METHODS: This population-based case-control analysis used data from the Korean National Health Insurance Service database (2004-2018). We compared 68,633 patients with new-onset IBD to matched controls (n = 343,165) using multivariable conditional logistic regression analysis. We also examined the dose-response relationship using non-linear regression analysis, and separately analysed childhood-onset IBD (aged ≤14 years) risk following early-life antibiotic exposure. RESULTS The mean age at diagnosis was 45.2 ± 16.8 years. Antibiotic prescriptions between 2 and 5 years before diagnosis significantly increased the odds of developing IBD (adjusted odds ratio [OR], 1.24; 95% confidence interval [CI]: 1.21-1.27). Additionally, sensitivity analysis revealed an elevated risk up to 9 years before diagnosis. Broad-spectrum antibiotics increased IBD risk, independent of gastroenteritis. A distinct dose-response relationship was observed irrespective of the IBD subtype and study population (all p < 0.001). Furthermore, antibiotic exposure within the first year of life was linked with the risk of childhood-onset IBD (OR, 1.51; 95% CI: 1.25-1.82). CONCLUSIONS Broad-spectrum antibiotics dose-dependently increased the risk for IBD in the Korean population. Our findings provide a fundamental epidemiological basis for identifying antibiotic use as a significant risk factor for IBD across different environmental backgrounds.
Collapse
Affiliation(s)
- Shin Ju Oh
- Department of Gastroenterology, Center for Crohn's and Colitis, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyo Jong Kim
- Department of Gastroenterology, Center for Crohn's and Colitis, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Chang Kyun Lee
- Department of Gastroenterology, Center for Crohn's and Colitis, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
45
|
Noble AJ, Nowak JK, Adams AT, Uhlig HH, Satsangi J. Defining Interactions Between the Genome, Epigenome, and the Environment in Inflammatory Bowel Disease: Progress and Prospects. Gastroenterology 2023; 165:44-60.e2. [PMID: 37062395 DOI: 10.1053/j.gastro.2023.03.238] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 04/18/2023]
Abstract
Recent advances in our understanding of the pathogenesis of inflammatory bowel disease (IBD) have highlighted the complex interplay between the genome, the epigenome, and the environment. Despite the exciting advances in genomics that have enabled the identification of over 200 susceptibility loci, these only account for a small proportion of the disease variance and the estimated heritability in IBD. It is likely that gene-environment (GxE) interactions contribute to "missing heritability" and these may act through epigenetic mechanisms. Several environmental factors, such as the microbiome, nutrition, and tobacco smoking, induce alterations in the epigenome of children and adults, which may impact disease susceptibility. Other mechanisms for GxE interactions are also directly pertinent in early life. We discuss a model in which environmental factors imprint disease risk in a window of susceptibility during infancy that may contribute to later disease onset, whereas other elements of the exposome act later in life and contribute directly to the pathogenesis and course of the disease. Understanding the mechanisms underlying GxE interactions may provide the basis for new therapeutic targets or preventative strategies for IBD.
Collapse
Affiliation(s)
- Alexandra J Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom.
| | - Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Alex T Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Department of Pediatrics, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
46
|
BASOGLU IA, KARAKOYUN B. Crohn’s disease: Etiology, pathogenesis and treatment strategies. MARMARA MEDICAL JOURNAL 2023; 36:249-254. [DOI: 10.5472/marumj.1307982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Crohn’s disease (CD), which can be localized in any part of the gastrointestinal tract, is a disease characterized by an irregular immune
response to normal and/or abnormal microbial antigens. Recent studies show many extensive data about the roles of genetic and
environmental factors, immune function, and gut microbiota in CD. Although, less invasive biomarkers are currently being developed,
the diagnosis of the disease is still based on the endoscopy and histological evaluation of biopsy samples. The most common symptoms
are diarrhea, abdominal pain, weight loss, and fatigue. Despite the improvements in the treatment methods in the last decade, there
is no definitive treatment since the etiology of CD is not known exactly. Therapeutic strategies focus on reducing inflammation and
symptoms, maintaining clinical remission, and improving quality of life.
Collapse
|
47
|
Abstract
Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8+ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8+ T cells are created equally and that the functions of individual CD8+ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8+ T cells and propose that the layered immune model be extended to the CD8+ T cell compartment.
Collapse
Affiliation(s)
- Cybelle Tabilas
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
48
|
Pantoja A, Sveum S, Frost S, Duran A, Burks J, Schernecke C, Feinberg M. New strategies to Reduce Unnecessary Antibiotic Use in the NICU: A Quality Improvement Initiative. Pediatr Qual Saf 2023; 8:e659. [PMID: 38571732 PMCID: PMC10990351 DOI: 10.1097/pq9.0000000000000659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/05/2023] [Indexed: 04/05/2024] Open
Abstract
Introduction Early-onset sepsis (EOS) and late-onset Sepsis (LOS) are common diagnoses entertained in sick newborns treated in neonatal intensive care units (NICUs), and antibiotics are the medications most prescribed in NICUs. Antibiotic stewardship programs have an important impact on limiting unnecessary antibiotic use. Methods Following the Model for Improvement, between 2/1/16 and 1/31/17, at a level 3 NICU, a multidisciplinary team implemented PDSA cycles to promote antibiotic stewardship practices for newborns at risk of EOS and LOS. The main goal was to decrease the antibiotic usage rate (AUR) safely. Primary strategies included discontinuing antibiotics within 24 hours of life if the newborn was stable, and the blood culture was negative for EOS and implementing an "antibiotic time-out" during rounds. Results For all newborns admitted to our NICU, the AUR decreased, for EOS from 137 to 32 days per 1000 patient days (77% reduction) and for LOS from 277 to 121 days per 1000 patient days (56% reduction). We demonstrated the sustainability of both EOS-AUR and LOS-AUR during the 2 years postcompletion of the intervention period. There were no adverse effects of reducing the AUR. Conclusion Interventions that reduce unnecessary antibiotic use in the NICU are safe and prevent excessive antibiotic exposure.
Collapse
Affiliation(s)
- Alfonso Pantoja
- From the Saint Joseph Hospital, Denver, Colo
- Colorado Permanente Medical Group, Denver, CO
| | - Scott Sveum
- From the Saint Joseph Hospital, Denver, Colo
- Vail Health, Frisco, CO
| | | | | | - Jeanne Burks
- From the Saint Joseph Hospital, Denver, Colo
- Envision Physician Services, Phoenix, AZ
| | - Christi Schernecke
- From the Saint Joseph Hospital, Denver, Colo
- Methodist Dallas Medical Center, Dallas TX
| | - Michelle Feinberg
- From the Saint Joseph Hospital, Denver, Colo
- Colorado Permanente Medical Group, Denver, CO
| |
Collapse
|
49
|
Kaur R, Kaur R, Varghese A, Garg N, Arora S. Antibiotics in Paediatrics: A Boon or a Bane? ANTI-INFECTIVE AGENTS 2023; 21. [DOI: 10.2174/2211352520666220822145139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 01/05/2025]
Abstract
Antibiotics play an essential role in antimicrobial therapy. Among all the medications
in children, the most commonly prescribed therapy is antibiotics and is currently the indispensable
means to cure transmissible diseases. Several categories of antibiotics have been introduced into
clinical practice to treat microbial infections. Reducing the unnecessary use of antibiotics is a
global need and priority. This article aims to provide better knowledge and understanding of the
impact of the early use of antibiotics. This article highlights the proper use of antibiotics in children,
detailing how early and inappropriate use of antibiotics affect the gut microbiome during
normal body development and consequently affect the metabolism due to diabetes mellitus, obesity,
and recurrence of infections, such as UTI. Several new antibiotics in their development stage,
newly marketed antibiotics, and some recalled and withdrawn from the market are also briefly
discussed in this article. This study will help future researchers in exploring the latest information
about antibiotics used in paediatrics.
Collapse
Affiliation(s)
- Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Rupinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Ashlin Varghese
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Nidhi Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| |
Collapse
|
50
|
Majer C, Lingel H, Arra A, Heuft HG, Bretschneider D, Balk S, Vogel K, Brunner-Weinzierl MC. PD-1/PD-L1 Control of Antigen-Specifically Activated CD4 T-Cells of Neonates. Int J Mol Sci 2023; 24:ijms24065662. [PMID: 36982735 PMCID: PMC10051326 DOI: 10.3390/ijms24065662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Newborns are highly susceptible to infections; however, the underlying mechanisms that regulate the anti-microbial T-helper cells shortly after birth remain incompletely understood. To address neonatal antigen-specific human T-cell responses against bacteria, Staphylococcus aureus (S. aureus) was used as a model pathogen and comparatively analyzed in terms of the polyclonal staphylococcal enterotoxin B (SEB) superantigen responses. Here, we report that neonatal CD4 T-cells perform activation-induced events upon S. aureus/APC-encounter including the expression of CD40L and PD-1, as well as the production of Th1 cytokines, concomitant to T-cell proliferation. The application of a multiple regression analysis revealed that the proliferation of neonatal T-helper cells was determined by sex, IL-2 receptor expression and the impact of the PD-1/PD-L1 blockade. Indeed, the treatment of S. aureus-activated neonatal T-helper cells with PD-1 and PD-L1 blocking antibodies revealed the specific regulation of the immediate neonatal T-cell responses with respect to the proliferation and frequencies of IFNγ producers, which resembled in part the response of adults’ memory T-cells. Intriguingly, the generation of multifunctional T-helper cells was regulated by the PD-1/PD-L1 axis exclusively in the neonatal CD4 T-cell lineage. Together, albeit missing memory T-cells in neonates, their unexperienced CD4 T-cells are well adapted to mount immediate and strong anti-bacterial responses that are tightly controlled by the PD-1/PD-L1 axis, thereby resembling the regulation of recalled memory T-cells of adults.
Collapse
Affiliation(s)
- Christiane Majer
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Aditya Arra
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Hans-Gert Heuft
- Institute of Transfusion Medicine and Immunohematology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | | | - Silke Balk
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Katrin Vogel
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Monika C. Brunner-Weinzierl
- Department of Experimental Pediatrics, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6724003
| |
Collapse
|