1
|
Nunez H, Nieto PA, Mars RA, Ghavami M, Sew Hoy C, Sukhum K. Early life gut microbiome and its impact on childhood health and chronic conditions. Gut Microbes 2025; 17:2463567. [PMID: 39916516 PMCID: PMC11810090 DOI: 10.1080/19490976.2025.2463567] [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: 09/23/2024] [Revised: 12/20/2024] [Accepted: 02/02/2025] [Indexed: 02/12/2025] Open
Abstract
The development of the gut microbiome is crucial to human health, particularly during the first three years of life. Given its role in immune development, disturbances in the establishment process of the gut microbiome may have long term consequences. This review summarizes evidence for these claims, highlighting compositional changes of the gut microbiome during this critical period of life as well as factors that affect gut microbiome development. Based on human and animal data, we conclude that the early-life microbiome is a determinant of long-term health, impacting physiological, metabolic, and immune processes. The early-life gut microbiome field faces challenges. Some of these challenges are technical, such as lack of standardized stool collection protocols, inconsistent DNA extraction methods, and outdated sequencing technologies. Other challenges are methodological: small sample sizes, lack of longitudinal studies, and poor control of confounding variables. To address these limitations, we advocate for more robust research methodologies to better understand the microbiome's role in health and disease. Improved methods will lead to more reliable microbiome studies and a deeper understanding of its impact on health outcomes.
Collapse
Affiliation(s)
- Harold Nunez
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
| | | | - Ruben A. Mars
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
2
|
Zhang D, Xie D, Qu Y, Mu D, Wang S. Digging deeper into necrotizing enterocolitis: bridging clinical, microbial, and molecular perspectives. Gut Microbes 2025; 17:2451071. [PMID: 39826099 DOI: 10.1080/19490976.2025.2451071] [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: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Necrotizing Enterocolitis (NEC) is a severe, life-threatening inflammatory condition of the gastrointestinal tract, especially affecting preterm infants. This review consolidates evidence from various biomedical disciplines to elucidate the complex pathogenesis of NEC, integrating insights from clinical, microbial, and molecular perspectives. It emphasizes the modulation of NEC-associated inflammatory pathways by probiotics and novel biologics, highlighting their therapeutic potential. We further critically examine dysbiotic alterations within the gut microbiota, with a particular focus on imbalances in bacterial and viral communities, which may contribute to the onset of NEC. The intricate interactions among toll-like receptor 4 (TLR4), microvascular integrity, immune activation, and the inflammatory milieu are meticulously summarized, offering a sophisticated understanding of NEC pathophysiology. This academic review aims to enhance the etiological comprehension of NEC, promote the development of targeted therapeutic interventions, and impart the significant impact of perinatal factors on the formulation of preventive and curative strategies for the disease.
Collapse
Affiliation(s)
- Deshuang Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dongke Xie
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Preidis GA. The neonatal gut microbiome in health and disease. Gut Microbes 2025; 17:2457499. [PMID: 39868670 PMCID: PMC11776465 DOI: 10.1080/19490976.2025.2457499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Affiliation(s)
- Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Yue F, Jiang M, Xu J, Ma J, Sun X, Huang J, Muratkhan M, Wang X, Lü X. Effect of pectinase addition in juice processing on the structural characteristics, immunological activity and in vitro and in vivo prebiotic properties of apple pomace pectic polysaccharides. Food Funct 2025. [PMID: 40260575 DOI: 10.1039/d5fo00354g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Apple pomace is a waste in fruit juice processing and is an important raw material for pectin extraction. The addition of pectinase will not only change the juice characteristics but also affect the apple pomace. However, the differences in the structure and function of pectin obtained from the pectinase-treated (TAPP) and untreated apple pomace (NTAPP) are unclear. In this paper, TAPP and NTAPP (APPs) were prepared using the subcritical-water method. Structural analysis showed that the APPs were acidic-pectin polysaccharides with low molecular weight (Mw) and a high esterification degree (DE), but TAPP had a lower Mw, DE, and galacturonic acid content and smoother surface. Immune activity detection demonstrated that NTAPP can stimulate macrophage proliferation, phagocytosis, and cytokine release by activating the TLR4/p-ERK/p-NFκB pathway, while TAPP activates the TLR4/p-NFκB to stimulate macrophage phagocytosis and the cytokine release. In vitro fermentation characteristics indicate that anaerobic fermentation using APPs as the sole carbon source can significantly promote the production of lactic acid and the short-chain fatty acids (SCFAs). Microbial diversity analysis revealed that the APPs exhibit prebiotic properties, but their effects on the gut microbiota composition differ: TAPP mainly promotes the enrichment of Akkermansia, while NTAPP primarily enhances the abundance of Faecalibaculum and Dubosiella. Finally, structure-function correlation analysis suggests that monosaccharide composition (particularly mannose) and molecular weight (Mw) are key factors influencing the gut microbiota composition, providing a research direction for future studies on their structure-activity relationships.
Collapse
Affiliation(s)
- Fangfang Yue
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Miao Jiang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jiaxin Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jingyi Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xin Sun
- Xijing Hospital, the Fourth Military Medical University, No. 127, Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Jihong Huang
- State Key Laboratory of Crop Stress Adaptation and Improvement, College of Agriculture, Henan University, Kaifeng 475004, China
| | - Marat Muratkhan
- Department of Food Technology and Processing Products, Technical Faculty, Saken Seifullin Kazakh Agrotechnical University, Nur-Sultan, Kazakhstan
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
5
|
Yu Z, Swift KA, Hedges MA, Theiss AL, Andres SF. Microscopic messengers: Extracellular vesicles shaping gastrointestinal health and disease. Physiol Rep 2025; 13:e70292. [PMID: 40165585 PMCID: PMC11959161 DOI: 10.14814/phy2.70292] [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: 11/25/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The field of extracellular vesicles (EVs) is advancing rapidly, and this review aims to synthesize the latest research connected to EVs and the gastrointestinal tract. We will address new and emerging roles for EVs derived from internal sources such as the pancreas and immune system and how these miniature messengers alter organismal health or the inflammatory response within the GI tract. We will examine what is known about external EVs from dietary and bacterial sources and the immense anti-inflammatory, immune-modulatory, and proliferative potential within these nano-sized information carriers. EV interactions with the intestinal and colonic epithelium and associated immune cells at homeostatic and disease states, such as necrotizing enterocolitis (NEC) and inflammatory bowel disease (IBD) will also be covered. We will discuss how EVs are being leveraged as therapeutics or for drug delivery and conclude with a series of unanswered questions in the field.
Collapse
Affiliation(s)
- Zhantao Yu
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Kevin A. Swift
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Madeline A. Hedges
- Department of Neonatology, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Arianne L. Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
- Rocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
| | - Sarah F. Andres
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| |
Collapse
|
6
|
Pell LG, Qamar H, Bassani DG, Heasley C, Funk C, Chen CY, Shawon J, O'Callaghan KM, Pullenayegum E, Hamer DH, Haque R, Kabir M, Ahmed T, O'Kelly C, Hossain MI, Khan AZ, Loutet MG, Islam MS, Morris SK, Shah PS, Sherman PM, Sultana S, Mahmud AA, Saha SK, Sarker SA, Roth DE. Neonatal administration of Lactiplantibacillus plantarum ATCC 202195 with or without fructooligosaccharide in Bangladesh: a placebo-controlled randomized trial. mSphere 2025; 10:e0103224. [PMID: 39992135 PMCID: PMC11934310 DOI: 10.1128/msphere.01032-24] [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: 12/09/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Lactiplantibacillus plantarum ATCC 202195 (LP202195) plus fructooligosaccharide (FOS) for 7 days was previously shown to colonize the infant intestine up to 6 months of age and reduced sepsis rates among young infants in rural India. In a phase 2 randomized controlled trial in Dhaka, Bangladesh (N = 519), neonatal administration of LP202195 for 1 or 7 days, with or without FOS, increased LP202195 stool abundance from 14 to 60 days of age, versus placebo. Abundance progressively declined in the post-administration period and did not persist beyond 2 months of age. FOS did not affect LP202195 abundance or its duration of persistence. All regimens were well-tolerated and safe. The absence of LP202195 colonization was inconsistent with results from a prior trial. Additional large-scale trials of LP202195 ± FOS are needed to establish its efficacy in infants who do not become LP202195-colonized. IMPORTANCE Among infants born in Dhaka, Bangladesh, a 7-day regimen of Lactiplantibacillus plantarum ATCC 202195 (LP202195) plus fructooligosaccharide (FOS) did not colonize the infant gastrointestinal (GI) tract. The absence of colonization is inconsistent with a prior study of the same synbiotic regimen in India, in which LP202195 was shown to persist in the infant GI tract for up to 6 months. Sustained LP202195 colonization was thought to be required for the probiotic to impart its beneficial impact on newborn sepsis. Therefore, additional trials are warranted to confirm the previously observed effects of LP202195 on infant clinical outcomes in the absence of LP202195 colonization. Moreover, since regimens of LP202195 that did not include FOS were indistinguishable from the synbiotic in terms of colonization, safety, and tolerability, future trials should assess the role of FOS for clinical efficacy; removing FOS would reduce costs, an important consideration for scale-up. CLINICAL TRIALS This study has been registered at ClinicalTrials.gov as NCT05180201.
Collapse
Affiliation(s)
- Lisa G. Pell
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Huma Qamar
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Diego G. Bassani
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Cole Heasley
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Celine Funk
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chun-Yuan Chen
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jakaria Shawon
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Karen M. O'Callaghan
- Department of Nutritional Sciences, King’s College London, London, United Kingdom
| | - Eleanor Pullenayegum
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Davidson H. Hamer
- Department of Global Health, Boston University School of Public Health and Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Rashidul Haque
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mamun Kabir
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Ciobha O'Kelly
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Md Iqbal Hossain
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Afreen Z. Khan
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Miranda G. Loutet
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Shaun K. Morris
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Prakesh S. Shah
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Philip M. Sherman
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shamima Sultana
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Abdullah Al Mahmud
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Shafiqul A. Sarker
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Daniel E. Roth
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
He Q, Li M, Diao H, Zheng Q, Li M, Zhu Q, Cui W. Association of Dietary Live Microbe Intake With Mortality: Results From the National Health and Nutrition Examination Survey, 1999-2018. J Acad Nutr Diet 2025:S2212-2672(25)00109-1. [PMID: 40147756 DOI: 10.1016/j.jand.2025.03.007] [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: 06/21/2024] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The association between dietary intake of live microbes and mortality remains unclear. OBJECTIVE This study aims to investigate the relationship between dietary live microbial intake and all-cause and cause-specific mortality among adults in the United States. DESIGN This is a cross-sectional study of adults aged 20 years or older who participated in the 1999-2018 National Health and Nutrition Examination Survey. PARTICIPANTS AND SETTING The study utilized data from adults aged 20 years and older with complete dietary and mortality data from the National Health and Nutrition Examination Survey from 1999 to 2018. MAIN OUTCOME MEASURES Deaths from any cause are defined as all-cause mortality. The International Statistical Classification of Diseases, 10th Revision, and the National Center for Health Statistics classifications of heart disease (054-064) and malignant neoplasms (019-043) were used to identify disease-specific causes of death. STATISTICAL ANALYSES PERFORMED Cox proportional hazard regression was utilized to examine the associations between the consumption of dietary live microbes and all-cause and cause-specific mortality. Restricted cubic spline regression modeling was used to assess potential linear associations between dietary live microorganism intake and mortality. In addition, stratified analyses and sensitivity analyses of the association of dietary live microorganism intake with all-cause and cardiovascular deaths were performed to validate the robustness of the results. RESULTS The study included 31 836 participants, of whom 4160 died, including 1109 cardiovascular deaths and 915 cancer deaths. The study found that consuming live microbes from the diet was linked to a lower rate of all-cause and cardiovascular mortality, respectively (hazard ratio 0.80, 95% CI 0.72 to 0.89; P < .001; hazard ratio 0.79, 95% CI 0.65 to 0.95; P = .014). However, there was no significant association observed between microbial intake and cancer mortality (hazard ratio 0.93, 95% CI 0.75 to 1.17; P = .545). Restricted cubic spline demonstrates a linear association between dietary live microorganism intake and all-cause and cardiovascular mortality (P < .001). Furthermore, sensitivity analyses indicated that a high intake of live dietary microorganisms was associated with a lower risk of all-cause mortality and cardiovascular mortality (P < .05). CONCLUSIONS The study found that consuming live microbes through diet was linked to a lower rate of all-cause and cardiovascular mortality but not cancer mortality.
Collapse
Affiliation(s)
- Qingzhen He
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Mingshuo Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Houze Diao
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Qingzhao Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Mingyuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Qing Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China.
| |
Collapse
|
8
|
Zheng L, Zhang J, Yang J, Wang Y, Zhang Y, Fang K, Wu J, Zheng M. Association of the use of nonfood prebiotics, probiotics, and synbiotics with total and cause-specific mortality: a prospective cohort study. Nutr J 2025; 24:45. [PMID: 40114150 PMCID: PMC11924732 DOI: 10.1186/s12937-025-01104-w] [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: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The use of nonfood prebiotics, probiotics, and synbiotics has approximately tripled in the last 20 years. It is necessary to examine the associations of these substances with all-cause and cause-specific mortality in a large prospective cohort. METHODS This study included 53,333 adults from the National Health and Nutrition Examination Survey 1999-2018. All participants answered questions on the use of dietary supplements and medications, including prebiotics, probiotics, and synbiotics. Death outcomes were determined by linkage to National Death Index records through 31 December 2019. Cox proportional hazards models were used to estimate hazard ratios (HR) and 95% confidence intervals (CI) for mortality from all causes, heart diseases, cancer, and other causes. RESULTS During a mean follow-up of 10.6 years, 9117 deaths were documented, including 2364 heart disease deaths, 1964 cancer deaths, and 4700 other causes deaths. Compared to nonusers, nonfood prebiotic, probiotic, and synbiotic users had a 59% (HR 0.41, 95% CI 0.30 to 0.56), 56% (HR 0.44, 95% CI 0.26 to 0.74), 49% (HR 0.51, 95% CI 0.31 to 0.83), and 64% (HR 0.36, 95% CI 0.23 to 0.59) for lower risk of all-cause, cancer, heart disease, and other causes mortality, respectively. Moreover, the inverse association of the use of prebiotics, probiotics, and synbiotics with mortality was stronger in female participants and participants without hypertension. CONCLUSION The use of nonfood prebiotics, probiotics, and synbiotics is significantly associated with lower all-cause mortality, as well as deaths from heart disease, cancer, and other causes.
Collapse
Affiliation(s)
- Luyan Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Jiaqi Zhang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Yanbo Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Yina Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Kailu Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Jie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China.
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
9
|
Xie Y, Chen Q, Shan D, Pan X, Hu Y. Unraveling the role of the gut microbiome in pregnancy disorders: insights and implications. Front Cell Infect Microbiol 2025; 15:1521754. [PMID: 40125520 PMCID: PMC11925892 DOI: 10.3389/fcimb.2025.1521754] [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: 11/02/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
The gut microbiota is the collective term for the microorganisms that reside in the human gut. In recent years, advances in sequencing technology and bioinformatics gradually revealed the role of gut microbiota in human health. Dramatic changes in the gut microbiota occur during pregnancy due to hormonal and dietary changes, and these changes have been associated with certain gestational diseases such as preeclampsia (PE) and gestational diabetes mellitus (GDM). Modulation of gut microbiota has also been proposed as a potential treatment for these gestational diseases. The present article aims to review current reports on the association between gut microbiota and gestational diseases, explore possible mechanisms, and discuss the potential of probiotics in gestational diseases. Uncovering the link between gut microbiota and gestational diseases could lead to a new therapeutic approach.
Collapse
Affiliation(s)
- Yupei Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Dan Shan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Xiongfei Pan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- West China Second University Hospital, Sichuan University, Shuangliu Institute of Women’s and Children’s Health, Shuangliu Maternal and Child Health Hospital, Chengdu, China
| | - Yayi Hu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| |
Collapse
|
10
|
Li F, Armet AM, Korpela K, Liu J, Quevedo RM, Asnicar F, Seethaler B, Rusnak TBS, Cole JL, Zhang Z, Zhao S, Wang X, Gagnon A, Deehan EC, Mota JF, Bakal JA, Greiner R, Knights D, Segata N, Bischoff SC, Mereu L, Haqq AM, Field CJ, Li L, Prado CM, Walter J. Cardiometabolic benefits of a non-industrialized-type diet are linked to gut microbiome modulation. Cell 2025; 188:1226-1247.e18. [PMID: 39855197 DOI: 10.1016/j.cell.2024.12.034] [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: 05/29/2024] [Revised: 10/24/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Industrialization adversely affects the gut microbiome and predisposes individuals to chronic non-communicable diseases. We tested a microbiome restoration strategy comprising a diet that recapitulated key characteristics of non-industrialized dietary patterns (restore diet) and a bacterium rarely found in industrialized microbiomes (Limosilactobacillus reuteri) in a randomized controlled feeding trial in healthy Canadian adults. The restore diet, despite reducing gut microbiome diversity, enhanced the persistence of L. reuteri strain from rural Papua New Guinea (PB-W1) and redressed several microbiome features altered by industrialization. The diet also beneficially altered microbiota-derived plasma metabolites implicated in the etiology of chronic non-communicable diseases. Considerable cardiometabolic benefits were observed independently of L. reuteri administration, several of which could be accurately predicted by baseline and diet-responsive microbiome features. The findings suggest that a dietary intervention targeted toward restoring the gut microbiome can improve host-microbiome interactions that likely underpin chronic pathologies, which can guide dietary recommendations and the development of therapeutic and nutritional strategies.
Collapse
Affiliation(s)
- Fuyong Li
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Animal Science and Technology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Anissa M Armet
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Katri Korpela
- Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Uusimaa, Finland
| | - Junhong Liu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Rodrigo Margain Quevedo
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Tianna B S Rusnak
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Janis L Cole
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Zhihong Zhang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Shuang Zhao
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Xiaohang Wang
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Adele Gagnon
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edward C Deehan
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - João F Mota
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; Faculty of Nutrition, Federal University of Goiás, Goiânia, Goiás 74605-080, Brazil
| | - Jeffrey A Bakal
- Division of General Internal Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Russell Greiner
- Department of Computing Science, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Machine Intelligence Institute, Edmonton, AB T5J 3B1, Canada
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Laurie Mereu
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Andrea M Haqq
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Liang Li
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada; Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Carla M Prado
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; School of Microbiology, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Medicine, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
11
|
Zimmermann P, Kurth S, Giannoukos S, Stocker M, Bokulich NA. NapBiome trial: Targeting gut microbiota to improve sleep rhythm and developmental and behavioural outcomes in early childhood in a birth cohort in Switzerland - a study protocol. BMJ Open 2025; 15:e092938. [PMID: 40032396 DOI: 10.1136/bmjopen-2024-092938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION The gut-brain axis plays a crucial role in the regulation and development of psychological and physical processes. The first year of life is a critical period for the development of the gut microbiome, which parallels important milestones in establishing sleep rhythm and brain development. Growing evidence suggests that the gut microbiome influences sleep, cognition and early neurodevelopment. For term-born and preterm-born infants, difficulties in sleep regulation may have consequences on health. Identifying effective interventions on the gut-brain axis in early life is likely to have long-term implications for the health and development of at-risk infants. METHODS AND ANALYSES In this multicentre, four-group, double-blinded, placebo (PLC)-controlled randomised trial with a factorial design, 120 preterm-born and 260 term-born infants will be included. The study will investigate whether the administration of daily synbiotics or PLC for a duration of 3 months improves sleep patterns and neurodevelopmental outcomes up to 2 years of age. The trial will also: (1) determine the association between gut microbiota, sleep patterns and health outcomes in children up to 2 years of age; and (2) leverage the interactions between gut microbiota, brain and sleep to develop new intervention strategies for at-risk infants. ETHICS AND DISSEMINATION The NapBiome trial has received ethical approval by the Committee of Northwestern and Central Switzerland and Canton Vaud, Switzerland (#2024-01681). Outcomes will be disseminated through publication and will be presented at scientific conferences. Metagenomic data will be shared through the European Nucleotide Archive. TRIAL REGISTRATION NUMBER The US National Institutes of Health NCT06396689.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Community Health and Department of Paediatrics, Fribourg Hospital, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Salome Kurth
- Department of Psychology, University of Fribourg, Fribourg, Switzerland
| | - Stamatios Giannoukos
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Martin Stocker
- Neonatology, Children's Hospital Lucerne, Lucerne, Switzerland
| | - Nicholas A Bokulich
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Pammi M, Preidis GA. How accurate are labels of probiotic products marketed to infants? Pediatr Res 2025:10.1038/s41390-025-03971-9. [PMID: 40025117 DOI: 10.1038/s41390-025-03971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Affiliation(s)
- Mohan Pammi
- Department of Pediatrics, Division of Neonatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.
| | - Geoffrey A Preidis
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Houston, TX, USA
| |
Collapse
|
13
|
Athalye-Jape G, Esvaran M, Patole S, Nathan EA, Doherty DA, Sim E, Chandrasekaran L, Kok C, Schuster S, Conway P. Effects of a live versus heat-inactivated probiotic Bifidobacterium spp in preterm infants: a randomised clinical trial. Arch Dis Child Fetal Neonatal Ed 2025; 110:177-184. [PMID: 39153842 DOI: 10.1136/archdischild-2023-326667] [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: 11/21/2023] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Heat-inactivated probiotics (HPs) may provide an effective alternative to live probiotics (P) by avoiding their risks (eg, probiotic sepsis) while retaining the benefits. We assessed the safety and efficacy of a HP in very preterm (VP: gestation <32 weeks) infants. METHODS VP infants were randomly allocated to receive a HP or P mixture (Bifidobacterium breve M-16V, Bifidobacterium longum subsp. infantis M-63, Bifidobacterium longum subsp. longum BB536, total 3×109 CFU/day) assuring blinding. Primary outcome was faecal calprotectin (FCP) levels were compared after 3 weeks of supplementation. Secondary outcomes included faecal microbiota and short chain fatty acid (SCFA) levels. RESULTS 86 VP infants were randomised to HP or P group (n=43 each). Total FCP and SCFA were comparable between HP and P groups within 7 days (T1) and between day 21 and 28 (T2) after supplementation. At T2, median (range) FCP was 75 (8-563) in the HP group and 80 (21-277) in the P group (p=0.71). Propionate was significantly raised in both groups, while butyrate was significantly raised in the HP group (all p<0.01). Bacterial richness and diversity increased but was comparable between HP and P (p>0.05). Beta diversity showed similar community structures in both groups (all p>0.05). Changes in faecal Actinobacteria, Bacteroidetes and Bifidobacteriacae levels were comparable in both groups at T1 and T2. There was no probiotic sepsis. CONCLUSIONS HP was safe and showed no significant difference in FCP as compared with a live probiotic. Adequately powered trials are needed to assess the effects of HP on clinically significant outcomes in preterm infants. TRIAL REGISTRATION NUMBER ACTRN12618000489291.
Collapse
Affiliation(s)
- Gayatri Athalye-Jape
- Neonatal Directorate, King Edward Memorial Hospital for Women Perth, Subiaco, Western Australia, Australia
- University of Western Australia, Perth, Western Australia, Australia
| | - Meera Esvaran
- University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Patole
- Neonatology, King Edward Memorial Hospital, Perth, Western Australia, Australia
- Neonatology, Centre for Neonatal Research and Education, Perth, Western Australia, Australia
| | - Elizabeth A Nathan
- Women and Infants Research Foundation, Western Australia, Australia, Australia
| | - Dorota A Doherty
- School of Women's and Infants' Health, University of Western Australia, Perth, Western Australia, Australia
| | - Edric Sim
- Nanyang Technological University, Singapore
| | | | - Chooi Kok
- Neonatal Directorate, King Edward Memorial Hospital for Women Perth, Subiaco, Western Australia, Australia
| | | | - Patricia Conway
- Centre for Marine Bio-Innovation, University of Newsouth Wales, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Alshaikh BN, Ting J, Lee S, Lemyre B, Wong J, Afifi J, Beltempo M, Shah PS. Effectiveness and Risks of Probiotics in Preterm Infants. Pediatrics 2025; 155:e2024069102. [PMID: 39933567 DOI: 10.1542/peds.2024-069102] [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] [Received: 09/09/2024] [Accepted: 10/21/2024] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVE To evaluate the effectiveness and risks of probiotics among infants born before 34 weeks' gestation and with a birth weight less than 1000 g. METHODS A population-based retrospective cohort study of infants born before 34 weeks' gestation and admitted to 33 Canadian Neonatal Network (CNN) units between January 1, 2016, and December 31, 2022. We excluded infants who were moribund on admission, died within the first 2 days, were admitted to CNN sites more than 2 days after birth, had major congenital anomalies, or never received enteral feeds. Logistic regression, propensity score-matched, and inverse probability of treatment weighting analyses were applied. RESULTS Among 32 667 eligible infants born before 34 weeks' gestation, 18 793 (57.5%) (median [IQR] gestational age, 29 [27-31] weeks) received probiotics, and 13 874 (42.5%) (median [IQR] gestational age, 31 [29-33] weeks) did not receive probiotics. In these infants, probiotics were associated with decreased mortality rates (adjusted odds ratio [aOR], 0.62; 98.3% CI, 0.53-0.73) but not decreased rates of necrotizing enterocolitis (NEC) (aOR, 0.92; 98.3% CI, 0.78-1.09) or late-onset sepsis (aOR, 0.90; 98.3% CI, 0.80-1.01). In 7401 infants with a birth weight less than 1000 g, probiotics were associated with decreased mortality rates (aOR, 0.58; 98.3% CI, 0.47-0.71) but not decreased NEC (aOR, 0.90; 98.3% CI. 0.71-1.13) or late-onset sepsis rates (aOR, 1.01; 98.3% CI, 0.86-1.18). Probiotic sepsis occurred in 27 (1.4/1000) infants born before 34 weeks' gestation and 20 (4/1000) infants with a birth weight less than 1000 g. Three infants with probiotic sepsis died, with probiotic sepsis deemed a possible cause in 2 cases. CONCLUSION Probiotics used in Canadian neonatal units were associated with decreased mortality in infants born before 34 weeks' gestation and with a birth weight less than 1000 g with limited effects on NEC and late-onset sepsis. Probiotic sepsis was rare.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Gastroenterology and Nutrition Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joseph Ting
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Seungwoo Lee
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Brigitte Lemyre
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan Wong
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jehier Afifi
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marc Beltempo
- Division of Neonatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Prakesh S Shah
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Farella I, Fortunato M, Martinelli D, De Carlo C, Sparapano E, Stolfa S, Romanelli F, De Laurentiis V, Martinotti S, Capozzi L, Castellana S, Parisi A, Latorre G. Lactobacillus rhamnosus Sepsis in a Preterm Infant Following Probiotic Administration: Challenges in Diagnosis. Microorganisms 2025; 13:265. [PMID: 40005631 PMCID: PMC11857594 DOI: 10.3390/microorganisms13020265] [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: 01/10/2025] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Probiotic administration has become common practice in neonatal intensive care units (NICUs) to prevent necrotizing enterocolitis (NEC) and promote gut health in preterm infants. While probiotics are generally considered safe, rare cases of probiotic-related sepsis have been reported. We present a case of Lactobacillus rhamnosus sepsis in a preterm infant, highlighting the challenges involved in its diagnosis. The infant developed symptoms of sepsis on the 13th day of probiotic treatment. Laboratory analyses, including MALDI-TOF, BioFire BCID2 panel, and whole-genome sequencing (WGS), helped confirm the diagnosis and the presence of Lactobacillus rhamnosus. In this case, accurately identifying the Lactobacillus rhamnosus strain proved challenging, as initial analyses using the Vitek 2 system yielded incorrect identifications. This highlights the limitations of automated systems in distinguishing closely related species, reinforcing the need for advanced molecular techniques to achieve precise strain identification and confirm a probiotic-related infection. Given these diagnostic complexities, it is crucial for clinicians to maintain a high index of suspicion for probiotic-related infections in cases of unexplained sepsis, as this awareness can prompt further diagnostic investigations to ensure accurate pathogen identification. The infant responded to ampicillin therapy, showing clinical improvement within 10 days and was discharged in good health at 67 days of life. This case underscores the importance of advanced molecular diagnostic methods to confirm probiotic-related infections and highlights the need for caution in administering probiotics to vulnerable populations, such as preterm infants. Clinicians must maintain a high index of suspicion for probiotic-associated sepsis in unexplained cases of infection and tailor antibiotic therapy based on susceptibility profiles. These findings emphasize the need for rigorous monitoring, appropriate probiotic strain selection, and optimized safety protocols in NICUs to mitigate potential risks.
Collapse
Affiliation(s)
- Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| | - Maria Fortunato
- Unit of Clinical Pathology and Microbiology, Miulli Regional General Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Domenico Martinelli
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| | - Carmela De Carlo
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Eleonora Sparapano
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Stefania Stolfa
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Federica Romanelli
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Vittoriana De Laurentiis
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Stefano Martinotti
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
- Unit of Clinical Pathology and Microbiology, Miulli Regional General Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Loredana Capozzi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Stefano Castellana
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Giuseppe Latorre
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| |
Collapse
|
16
|
Quigley EMM, Shanahan F. Probiotics in Health Care: A Critical Appraisal. Annu Rev Med 2025; 76:129-141. [PMID: 39527719 DOI: 10.1146/annurev-med-042423-042315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Consumption of probiotic products continues to increase, perhaps driven by an interest in gut health. However, the field is filled with controversy, inconsistencies, misuse of terminology, and poor communication. While the probiotic concept is biologically plausible and in some cases mechanistically well established, extrapolation of preclinical results to humans has seldom been proven in well-conducted clinical trials. With noteworthy exceptions, clinical guidance has often been derived not from large, adequately powered clinical trials but rather from comparisons of disparate, small studies with insufficient power to identify the optimal strain. The separation of probiotics from live biotherapeutic products has brought some clarity from a regulatory perspective, but in both cases, consumers should expect scientific rigor and strong supporting evidence for health claims.
Collapse
Affiliation(s)
- Eamonn M M Quigley
- Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Fergus Shanahan
- Department of Medicine and Alimentary Pharmabiotic Centre, Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland;
| |
Collapse
|
17
|
Preidis GA, Berrington JE. Probiotics for preterms: sharing complex decision-making. Nat Rev Gastroenterol Hepatol 2025; 22:5-6. [PMID: 39496887 DOI: 10.1038/s41575-024-01009-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Affiliation(s)
- Geoffrey A Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.
| | - Janet E Berrington
- Newcastle Neonatal Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
Bi BC, Yang HY, Su JY, Deng L. Analysis of pregnancy and neonatal outcomes in 100 pregnant women with Rh-negative blood type. BMC Pregnancy Childbirth 2024; 24:815. [PMID: 39695493 DOI: 10.1186/s12884-024-06981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND This study aimed to explore variations in prenatal care, delivery methods, influencing factors, and neonatal outcomes among Rh-negative pregnant women, so as to improve pregnancy healthcare for this demographic, raise the quality of maternal-fetal management, and safeguard the health of both mother and infant. METHODS This study included 200 women who received routine prenatal care, exhibited no other pregnancy complications, and were admitted for delivery. They were divided into an observation group (100 Rh-negative blood type) and a control group (100 Rh-positive blood type). The study examined differences in pregnancy management, clinical characteristics and pregnancy outcomes between the two groups. RESULTS The results indicated that singleton pregnancies in Rh-negative mothers are associated with significantly higher rates of postpartum blood loss (305.1 ± 183.8 vs. 246.1 ± 84.9 mL, P = 0.004), neonatal hyperbilirubinemia (39% vs. 23%, P = 0.014), low birth weight (11% vs. 2%, P = 0.01), and NICU admission (30% vs. 18%, P = 0.046) compared to the control group. Among Rh-negative mothers, subgroup analysis by ethnicity revealed a higher incidence of fetal distress in the other ethnic groups compared to the Han and Zhuang groups (16.7%, 0, 6.5%, respectively, P = 0.025). Subgroup analysis based on ABO blood type within Rh-negative mothers did not show any statistical significance in various outcomes (all P > 0.05). Infants with neonatal hyperbilirubinemia born to Rh-negative mothers experienced a quicker resolution of hyperbilirubinemia compared to those whose mothers did not receive intramuscular anti-D immunoglobulin [1.0 (1.0, 1.5) vs. 5.0 (1.5, 10.0), P = 0.002]. CONCLUSIONS The Rh-negative blood type is linked to higher risks of neonatal hyperbilirubinemia, low birth weight, and increased postpartum hemorrhage, resulting in detrimental pregnancy outcomes. Administering anti-D immunoglobulin speeds up the resolution of neonatal hyperbilirubinemia. Thus, prudent and efficient use of anti-D immunoglobulin can mitigate adverse outcomes for both mothers and newborns.
Collapse
Affiliation(s)
- Bing-Cai Bi
- Department of Obstetrics, Maternal and Child, Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530002, Guangxi, China
| | - Hong-Yan Yang
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Jun-You Su
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| | - Li Deng
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| |
Collapse
|
19
|
Das A, Fleming P. Evaluating probiotic mechanisms may help inform optimum strain selection for use in clinical trials. Pediatr Res 2024; 96:1537-1538. [PMID: 39014243 DOI: 10.1038/s41390-024-03395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/30/2024] [Accepted: 05/22/2024] [Indexed: 07/18/2024]
Affiliation(s)
- Abhishek Das
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
| | - Paul Fleming
- Department of Neonatology, Homerton Healthcare NHS Foundation Trust, London, UK.
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
20
|
Rühle J, Schwarz J, Dietz S, Rückle X, Schoppmeier U, Lajqi T, Poets CF, Gille C, Köstlin-Gille N. Impact of perinatal administration of probiotics on immune cell composition in neonatal mice. Pediatr Res 2024; 96:1645-1654. [PMID: 38278847 PMCID: PMC11772233 DOI: 10.1038/s41390-024-03029-2] [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] [Received: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Newborns and especially preterm infants are much more susceptible to infections than adults. The pathogens causing infections in newborns are often detectable in the intestinal flora of affected children even before disease onset. Therefore, it seems reasonable to prevent dysbiosis in newborns and preterm infants. An approach followed in many neonatal intensive care units (NICUs) is to prevent infections in preterm infants with probiotics however their mechanisms of action of probiotics are incompletely understood. Here, we investigated the effect of perinatal probiotic exposure on immune cells in newborn mice. METHODS Pregnant mice were orally treated with a combination of Lactobacillus acidophilus and Bifidobacterium bifidum (Infloran®) from mid-pregnancy until the offspring were harvested. Immune cell composition in organs of the offspring were analyzed by flow cytometry. RESULTS Perinatal probiotic exposure had profound effects on immune cell composition in the intestine, liver and lungs of newborn mice with reduction of myeloid and B cells and induction of T cells in the probiotic treated animals' organs at weaning. Furthermore, probiotic exposure had an effect on T cell development in the thymus. CONCLUSION Our results contribute to a better understanding of the interaction of probiotics with the developing immune system. IMPACT probiotics have profound effects on immune cell composition in intestines, livers and lungs of newborn mice. probiotics modulate T cell development in thymus of newborn mice. effects of probiotics on neonatal immune cells are particularly relevant in transition phases of the microbiome. our results contribute to a better understanding of the mechanisms of action of probiotics in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Xenia Rückle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Ulrich Schoppmeier
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| |
Collapse
|
21
|
Lee Him R, Rehman S, Sihota D, Yasin R, Azhar M, Masroor T, Naseem HA, Masood L, Hanif S, Harrison L, Vaivada T, Sankar MJ, Dramowski A, Coffin SE, Hamer DH, Bhutta ZA. Prevention and Treatment of Neonatal Infections in Facility and Community Settings of Low- and Middle-Income Countries: A Descriptive Review. Neonatology 2024; 122:173-208. [PMID: 39532080 PMCID: PMC11875423 DOI: 10.1159/000541871] [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: 04/09/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION We present a robust and up-to-date synthesis of evidence on the effectiveness of interventions to prevent and treat newborn infections in low- and middle-income countries (LMICs). Newborn infection prevention interventions included strategies to reduce antimicrobial resistance (AMR), prevention of healthcare-associated infections (HAIs), clean birth kits (CBKs), chlorhexidine cleansing, topical emollients, and probiotic and synbiotic supplementation. Interventions to treat suspected neonatal infections included prophylactic systemic antifungal agents and community-based antibiotic delivery for possible serious bacterial infections (PSBIs). METHODS A descriptive review combining different methodological approaches was conducted. To provide the most suitable recommendations for real-world implementation, our analyses considered the impact of these interventions within three distinct health settings: facility, mixed, and community. RESULTS In facility settings, the strongest evidence supported the implementation of multimodal stewardship interventions for AMR reduction and device-associated infection prevention bundles for HAI prevention. Emollients in preterm newborns reduced the risk of invasive infection compared to routine skin care. Probiotics in preterm newborns reduced neonatal mortality, invasive infection, and necrotizing enterocolitis (NEC) risks compared to standard care or placebo. There was insufficient evidence for synbiotics and prophylactic systemic antifungals in LMICs. In mixed settings, CBKs reduced neonatal mortality risk compared to standard care. In community settings, chlorhexidine umbilical cord cleansing reduced omphalitis risk compared to dry cord care. For the treatment of PSBIs, purely domiciliary-based antibiotic delivery reduced the risk of all-cause neonatal mortality when compared to the standard hospital referral. CONCLUSION Strategies for preventing HAIs and reducing AMR in healthcare facilities should be multimodal, and strategy selection should consider the feasibility of integration within existing newborn care programs. Probiotics are effective for facility-based use in preterm newborns; however, the establishment of high-quality, cost-effective mass production of standardized formulations is needed. Chlorhexidine cord cleansing is effective in community settings to prevent omphalitis in contexts where unhygienic cord applications are prevalent. Community-based antibiotic delivery of simplified regimens for PSBIs is a safe alternative when hospital-based care in LMICs is not possible or is declined by parents. More randomized trial evidence is needed to establish the effectiveness of CBKs, emollients, synbiotics, and prophylactic systemic antifungals in LMICs. INTRODUCTION We present a robust and up-to-date synthesis of evidence on the effectiveness of interventions to prevent and treat newborn infections in low- and middle-income countries (LMICs). Newborn infection prevention interventions included strategies to reduce antimicrobial resistance (AMR), prevention of healthcare-associated infections (HAIs), clean birth kits (CBKs), chlorhexidine cleansing, topical emollients, and probiotic and synbiotic supplementation. Interventions to treat suspected neonatal infections included prophylactic systemic antifungal agents and community-based antibiotic delivery for possible serious bacterial infections (PSBIs). METHODS A descriptive review combining different methodological approaches was conducted. To provide the most suitable recommendations for real-world implementation, our analyses considered the impact of these interventions within three distinct health settings: facility, mixed, and community. RESULTS In facility settings, the strongest evidence supported the implementation of multimodal stewardship interventions for AMR reduction and device-associated infection prevention bundles for HAI prevention. Emollients in preterm newborns reduced the risk of invasive infection compared to routine skin care. Probiotics in preterm newborns reduced neonatal mortality, invasive infection, and necrotizing enterocolitis (NEC) risks compared to standard care or placebo. There was insufficient evidence for synbiotics and prophylactic systemic antifungals in LMICs. In mixed settings, CBKs reduced neonatal mortality risk compared to standard care. In community settings, chlorhexidine umbilical cord cleansing reduced omphalitis risk compared to dry cord care. For the treatment of PSBIs, purely domiciliary-based antibiotic delivery reduced the risk of all-cause neonatal mortality when compared to the standard hospital referral. CONCLUSION Strategies for preventing HAIs and reducing AMR in healthcare facilities should be multimodal, and strategy selection should consider the feasibility of integration within existing newborn care programs. Probiotics are effective for facility-based use in preterm newborns; however, the establishment of high-quality, cost-effective mass production of standardized formulations is needed. Chlorhexidine cord cleansing is effective in community settings to prevent omphalitis in contexts where unhygienic cord applications are prevalent. Community-based antibiotic delivery of simplified regimens for PSBIs is a safe alternative when hospital-based care in LMICs is not possible or is declined by parents. More randomized trial evidence is needed to establish the effectiveness of CBKs, emollients, synbiotics, and prophylactic systemic antifungals in LMICs.
Collapse
Affiliation(s)
- Rachel Lee Him
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Sarah Rehman
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Davneet Sihota
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Rahima Yasin
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Maha Azhar
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Taleaa Masroor
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Hamna Amir Naseem
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Laiba Masood
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sawera Hanif
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Leila Harrison
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Tyler Vaivada
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
| | - M. Jeeva Sankar
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Angela Dramowski
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Susan E. Coffin
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Davidson H. Hamer
- Department of Global Health, Boston University School of Public Health, Boston, MA, USA
- Section of Infectious Diseases, Department of Medicine, Boston University Avedisian and Chobanian School of Medicine, Boston, MA, USA
- Center on Emerging Infectious Diseases, Boston University, Boston, MA, USA
| | - Zulfiqar A. Bhutta
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
22
|
Xuan H, Umar S, Zhong C, Yu W, Ahmed I, Wheatley JL, Sampath V, Chavez-Bueno S. Lactobacillus rhamnosus modulates murine neonatal gut microbiota and inflammation caused by pathogenic Escherichia coli. BMC Microbiol 2024; 24:452. [PMID: 39506682 PMCID: PMC11539828 DOI: 10.1186/s12866-024-03598-6] [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: 06/19/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Pathogenic Escherichia coli strains produce neonatal septicemia after colonizing the neonatal gut. While the probiotic Lactobacillus rhamnosus GG (LGG) effectively reduces neonatal sepsis, LGG's effects on the neonatal intestinal microbiota alterations and inflammation triggered by E. coli are incompletely understood. We hypothesized that LGG significantly modulates the specific neonatal gut microbial populations changes and the inflammatory response elicited by the enteral introduction of septicemia-producing E. coli. To test this hypothesis, newborn rats were pretreated orally with LGG or placebo prior to infection with the neonatal E. coli septicemia clinical isolate SCB34. Amplicon 16S rRNA gene sequencing was performed on intestinal samples. Intestinal injury and expression of inflammatory mediators and apoptosis were determined. RESULTS Alpha diversity of gut microbiota was greater in SCB34-infected pups in comparison to sham-infected pups, these changes were not modified by LGG pretreatment. Beta diversity analyses also showed differences between SCB34-infected vs. uninfected pups. LGG pretreatment before SCB34 infection did not result in significant beta diversity changes compared to placebo. Moreover, individual genera and species abundance analyses by linear discriminant analysis effect size (LEfSe) showed significant changes in Gram-negative, Gram-positive, and anaerobic populations resulting from LGG pretreatment and SCB34 infection. LGG significantly suppressed the expression of inflammatory cytokines but did not attenuate SCB34-induced apoptosis or histologic injury. CONCLUSIONS LGG modulates clinically significant microbiota features and inflammation triggered by pathogenic E. coli intestinal infection shortly after birth. This new knowledge can potentially be harnessed to design novel interventions against gut-derived neonatal sepsis.
Collapse
Affiliation(s)
- Hao Xuan
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Wei Yu
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Ishfaq Ahmed
- Department of Math, Science and Computer Technology, Kansas City Kansas Community College, Kansas City, KS, USA
| | - Joshua L Wheatley
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Susana Chavez-Bueno
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA.
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| |
Collapse
|
23
|
More K, Hanumantharaju A, Amrit A, Nimbalkar SM, Patole S. Use of Probiotics for Preventing Necrotizing Enterocolitis in Preterm Infants: A Survey of Current Practices Among Indian Neonatologists. Cureus 2024; 16:e73923. [PMID: 39697933 PMCID: PMC11654888 DOI: 10.7759/cureus.73923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE Probiotics are known to reduce the risk of necrotizing enterocolitis (NEC≥ Stage II) significantly, as well as all-cause mortality, late-onset sepsis (LOS), and feeding intolerance in preterm infants. Probiotics have been reported to have comparable benefits in high- and low-middle-income countries (LMICs). We aimed to assess the current practices of neonatologists in India for using probiotics in preterm infants. MATERIAL AND METHODS A questionnaire created using Survey Monkey's web-based tool was sent to neonatologists in India. Survey forms automatically converted responses into Excel files (Microsoft® Corp., Redmond, WA). Data were analyzed using SPSS (IBM Corp., Armonk, NY). RESULTS A total of 615 responses were received from various neonatal intensive care units (NICUs) in India (Level I: 43 (7%), II: 124 (20.8%), III: 448 (72.8%)). Around 431 (70%) of the units had either National Neonatology Forum (NNF) accreditation or IAP fellowships or were affiliated with private or government medical colleges. The remaining 184 (30%) were in private setups. Routine probiotic supplementation (RPS) was provided in 241 (39.1%) of the responding units; 179 (48%) quoted inadequate evidence as the reason for not providing RPS, 125 (33.43%) quoted difficulty in sourcing safe and effective products, whereas others were concerned about adverse effects. Most centers provided RPS for preterm infants <32 weeks and 1500 g at birth. The clinical practice was influenced by the judgment of the attending clinician. Significant variation was noticed in the protocol for RPS. CONCLUSION Findings of the survey suggest that approximately 39% of the participating neonatologists in India currently offer RPS for preterm infants. A significant variation exists in the selection of probiotic strains, products, dose, and duration of supplementation. Despite limitations, our findings are useful in guiding clinical practice and further research to optimize the safety and efficacy of RPS for preterm infants.
Collapse
Affiliation(s)
- Kiran More
- Neonatology, MRR Children's Hospital, Thane, IND
| | - Anil Hanumantharaju
- Neonatology, Pramukhswami Medical College, Bhaikaka University, Karamsad, IND
| | - Astha Amrit
- Neonatology, Mount Sinai Hospital, Toronto, CAN
| | - Somashekhar M Nimbalkar
- Central Research Services, Bhaikaka University, Karamsad, IND
- Pediatrics, Pramukhswami Medical College, Karamsad, IND
| | - Sanjay Patole
- Neonatology, King Edward Memorial Hospital, Perth, AUS
| |
Collapse
|
24
|
Ananthan A, Balasubramanian H, Rath C, Muthusamy S, Rao S, Patole S. Lactobacillus rhamnosus GG as a probiotic for preterm infants: a strain specific systematic review and meta-analysis. Eur J Clin Nutr 2024; 78:830-846. [PMID: 39060543 DOI: 10.1038/s41430-024-01474-0] [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: 03/26/2023] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Lactobacillus rhamnosus GG (LGG) is a widely used and extensively researched probiotic. Probiotic effects are considered to be strain specific. We aimed to comprehensively assess the strain-specific effects of LGG in preterm infants. A systematic review of RCTs and non-RCTs to evaluate the effect of LGG in preterm infants. We followed the Cochrane methodology, and preferred reporting items for systematic reviews (PRISMA) statement for conducting and reporting this review. We searched the Cochrane central register of controlled trials, PubMed, EMBASE and CINAHL databases till December 2023. The review was registered in PROSPERO 2022 CRD42022324933. Meta-analysis of data from RCTs that used LGG as the sole probiotic showed significantly lower risk of NEC ≥Stage II [5 RCTs, n = 851, RR:0.50 (95% CI: 0.26, 0.93), P = 0.03] in the LGG group. There was no significant difference in the risk of LOS [7 RCTs, n = 1037, RR:1.08 (95% CI 0.84, 1.39), P = 0.55], mortality [3 RCTs, n = 207, RR: 0.99 (95% CI: 0.42, 2.33), P = 0.99], time to reach full feeds [2 RCTs, n = 19, SMD = 0.11 days (95% CI: -0.22, 0.45), P = 0.51] and duration of hospital stay [3 RCTs, n = 293, SMD: -0.14 days (95% CI: -0.37 to 0.09), P = 0.23]. Meta-analysis of data from non-RCTs showed no significant effect of LGG on NEC, LOS, and mortality. RCTs showed beneficial effects of LGG when used as the sole probiotic in reducing the risk of NEC, whereas observational studies did not. Strain-specific systematic review of LGG provides important data for guiding research and clinical practice.
Collapse
Affiliation(s)
- Anitha Ananthan
- Department of Neonatology, Seth GS Medical College and King Edward Memorial Hospital, Mumbai, India.
| | | | - Chandra Rath
- Department of Neonatology, Joondalup Health Campus, Perth, WA, Australia
| | | | - Shripada Rao
- Department of Neonatology, Joondalup Health Campus, Perth, WA, Australia
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Sanjay Patole
- School of Medicine, University of Western Australia, Perth, WA, Australia
- Neonatal Directorate, King Edward Memorial Hospital for Women, Perth, WA, Australia
| |
Collapse
|
25
|
Hu J, Yao Q, Zhao L. Evidences and perspectives on the association between gut microbiota and sepsis: A bibliometric analysis from 2003 to 2023. Heliyon 2024; 10:e37921. [PMID: 39315201 PMCID: PMC11417584 DOI: 10.1016/j.heliyon.2024.e37921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Background In the last two decades, the role of the gut microbiome in the development, maintenance, and outcome of sepsis has received increased attention; however, few descriptive studies exist on its research focus, priorities, and future prospects. This study aimed to identify the current state, evolution, and emerging trends in the field of gut microbiota and sepsis using bibliometric analysis. Methods All publications on sepsis and gut microbiota were retrieved from the Web of Science Core Collection and included in this study. VOSviewer, CiteSpace, and the Web of Science online analysis platform were used to visualize trends based on publication country, institution, author, journal, and keywords. Results A total of 1,882 articles on sepsis-related gut microbiota were screened, mainly from 95 countries or regions and 2,581 institutions. The United States and China contributed the most to this research field, with 521 (27.683 %) and 376 (19.979 %) articles, respectively. Scientists from the University of California were the most prolific, publishing 63 (3.348 %) articles. Cani PD published papers with the highest H-index, establishing himself as a leader in the field. The most publications were published in the journals "Nutrients" and "PLOS One." The journals with the most co-citations were "PLOS One," "Nature," and "Gut." The most used keywords were prebiotics, gut microbiota, and sepsis. The keyword burst research analysis revealed that research on treatment strategies based on the intestinal microbiota, intestine-liver axis, and regulatory mechanisms of bacterial metabolites are currently hot directions. Conclusion This study presents a global overview of the current state and potential trends in the field of sepsis-related gut microbiota. This study identified hot research sub-directions and new trends through comparison and analysis, which will aid in the development of this field.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou City, 310003, China
| | - Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Linjun Zhao
- Department of Emergency, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Rd, Hangzhou City, 310006, China
| |
Collapse
|
26
|
Cuna A, Kumar N, Sampath V. Understanding necrotizing enterocolitis endotypes and acquired intestinal injury phenotypes from a historical and artificial intelligence perspective. Front Pediatr 2024; 12:1432808. [PMID: 39398415 PMCID: PMC11466774 DOI: 10.3389/fped.2024.1432808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
Necrotizing enterocolitis (NEC) remains a devastating disease in preterm and term neonates. Despite significant progress made in understanding NEC pathogenesis over the last 50 years, the inability of current definitions to discriminate the various pathophysiological processes underlying NEC has led to an umbrella term that limits clinical and research progress. In this mini review, we provide a historical perspective on how NEC definitions and pathogenesis have evolved to our current understanding of NEC endotypes. We also discuss how artificial intelligence-based approaches are influencing our knowledge of risk-factors, classification and prognosis of NEC and other neonatal intestinal injury phenotypes.
Collapse
Affiliation(s)
- Alain Cuna
- Division of Neonatology, Children’s Mercy Kansas City, Kansas City, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Navin Kumar
- Division of Neonatology, Hurley Medical Center, Flint, MI, United States
| | - Venkatesh Sampath
- Division of Neonatology, Children’s Mercy Kansas City, Kansas City, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
27
|
Gonia S, Heisel T, Miller N, Haapala J, Harnack L, Georgieff MK, Fields DA, Knights D, Jacobs K, Seburg E, Demerath EW, Gale CA, Swanson MH. Maternal oral probiotic use is associated with decreased breastmilk inflammatory markers, infant fecal microbiome variation, and altered recognition memory responses in infants-a pilot observational study. Front Nutr 2024; 11:1456111. [PMID: 39385777 PMCID: PMC11462058 DOI: 10.3389/fnut.2024.1456111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Early life gut microbiomes are important for brain and immune system development in animal models. Probiotic use has been proposed as a strategy to promote health via modulation of microbiomes. In this observational study, we explore if early life exposure to probiotics via the mother during pregnancy and lactation, is associated with decreased inflammation in breastmilk, maternal and infant microbiome variation, and altered infant neurodevelopmental features. Methods Exclusively breastfeeding mother-infant dyads were recruited as part of the "Mothers and Infants Linked for Healthy Growth (MILk) Study." Probiotic comparison groups were defined by exposure to maternal probiotics (NO/YES) and by timing of probiotic exposure (prenatal, postnatal, total). C-reactive protein (CRP) and IL-6 levels were determined in breastmilk by immunoassays, and microbiomes were characterized from 1-month milk and from 1- and 6-month infant feces by 16S rDNA sequencing. Infant brain function was profiled via electroencephalogram (EEG); we assessed recognition memory using event-related potential (ERP) responses to familiar and novel auditory (1 month) and visual (6 months) stimuli. Statistical comparisons of study outcomes between probiotic groups were performed using permutational analysis of variance (PERMANOVA) (microbiome) and linear models (all other study outcomes), including relevant covariables as indicated. Results We observed associations between probiotic exposure and lower breastmilk CRP and IL-6 levels, and infant gut microbiome variation at 1- and 6-months of age (including higher abundances of Bifidobacteria and Lactobacillus). In addition, maternal probiotic exposure was associated with differences in infant ERP features at 6-months of age. Specifically, infants who were exposed to postnatal maternal probiotics (between the 1- and 6-month study visits) via breastfeeding/breastmilk, had larger differential responses between familiar and novel visual stimuli with respect to the late slow wave component of the EEG, which may indicate greater memory updating potential. The milk of mothers of this subgroup of infants had lower IL-6 levels and infants had different 6-month fecal microbiomes as compared to those in the "NO" maternal probiotics group. Discussion These results support continued research into "Microbiota-Gut-Brain" connections during early life and the role of pre- and postnatal probiotics in mothers to promote healthy microbiome-associated outcomes in infants.
Collapse
Affiliation(s)
- Sara Gonia
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Timothy Heisel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Neely Miller
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | - Jacob Haapala
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Lisa Harnack
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Michael K. Georgieff
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - David A. Fields
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Katherine Jacobs
- Division of Maternal-Fetal Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Elisabeth Seburg
- Pregnancy and Child Health Research Center, HealthPartners Institute, Bloomington, MN, United States
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Cheryl A. Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Marie H. Swanson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
28
|
Wang H, Chen D, Li H, Fu C, Fang L, Wang R, Xu J. Bifidobacterium regulates premature infant gut metabolites, reducing serum inflammatory factors: a randomised controlled trial. Pediatr Res 2024:10.1038/s41390-024-03552-2. [PMID: 39271904 DOI: 10.1038/s41390-024-03552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Analyse the effects of Bifidobacterium BB-12 on intestinal metabolites and serum inflammatory factors in premature infants. METHODS 71 premature infants at gestational age of ≤32 weeks were randomly divided into the probiotic (n = 36) and control (n = 35) groups. Faecal and blood samples were collected from the two groups of premature infants at the 2nd and 4th week of life for intestinal metabolite detection and assessment of the level of the serum inflammatory markers TLR4, NF- κ B, IL-1β, and TNF- α. RESULTS Compared to the control group, the probiotic group contained more amino acids, these elements were enriched on multiple amino acid metabolic pathways, and the probiotic group showed significantly lower levels of the serum inflammatory markers TLR4, NF-κB, IL-1β, and TNF-α. Finally, the probiotic group showed a lower incidence of feeding intolerance. CONCLUSIONS The administration of Bifidobacterium BB-12 is associated with increasing the levels of glutamine, glutamic acid, and kynurenine in the gut of premature infants, and associated with reducing the levels of TLR4 and NF-κB in the serum, further decreasing the secretion of the pro-inflammatory factors IL-1β and TNF-α, and alleviating systemic inflammatory reactions, thereby reducing the incidence of feeding intolerance. IMPACT 1. The use of Bifidobacterium BB-12 in premature infants can increase the levels of amino acids in the intestine. 2. Increases in Bifidobacterium BB-12 may decrease the serum levels of TLR4, NF-κB, IL-1β, and TNF-α. 3. Kynurenine may improve the prognosis of preterm infants by reducing inflammation. 4. Bifidobacterium BB-12 may improve the feeding tolerance of premature infants, thus reducing the incidence of feeding intolerance.
Collapse
Affiliation(s)
- He Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Dongmei Chen
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Huamei Li
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Chunyan Fu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Lingyu Fang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Ruiquan Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Jinglin Xu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China.
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China.
| |
Collapse
|
29
|
Wu WH, Chiang MC, Fu RH, Lai MY, Wu IH, Lien R, Lee CC. Impact of Clinical Use of Probiotics on Preterm-Related Outcomes in Infants with Extremely Low Birth Weight. Nutrients 2024; 16:2995. [PMID: 39275310 PMCID: PMC11397160 DOI: 10.3390/nu16172995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/12/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
Background: Preterm birth significantly contributes to mortality and morbidities, with recent studies linking these issues to gut microbiota imbalances. Probiotic supplementation shows promise in mitigating adverse outcomes in preterm infants, but optimal timing and guidelines remain unclear. This study assesses the benefits of probiotic supplementation for preterm infants without consistent guidelines. Methods: This retrospective study examined extremely low-birth-weight (ELBW) infants in neonatal intensive care units from 2017 to 2021. Mortality and preterm-related outcomes were compared between infants receiving probiotics and those not. Subgroup analyses based on probiotic initiation timing were conducted: early (≤14 days), late (>14 days), and non-probiotic groups. Results: The study included 330 ELBW infants: 206 received probiotics (60 early, 146 late), while 124 did not. Probiotic supplementation was associated with lower overall mortality (adjusted OR 0.22, 95% CI 0.09-0.48) and decreased mortality from necrotizing enterocolitis (NEC) or late-onset sepsis (LOS) (adjusted OR 0.12, 95% CI 0.03-0.45). Early probiotics reduced overall mortality, NEC/LOS-related mortality, and NEC/LOS-unrelated mortality. Late probiotics decreased overall mortality and NEC/LOS-related mortality. Early probiotic use also expedited full enteral feeding achievement. Conclusions: Probiotic supplementation reduces mortality and improves feeding tolerance in preterm infants. Establishing guidelines for probiotic use in this population is crucial.
Collapse
Affiliation(s)
- Wei-Hung Wu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ren-Huei Fu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Mei-Yin Lai
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - I-Hsyuan Wu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Reyin Lien
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
30
|
Zeng Q, Zeng L, Yu X, Yuan X, Ma W, Song Z, Chen D. Clinical value of prokineticin 2 in the diagnosis of neonatal necrotizing enterocolitis. Biomarkers 2024; 29:361-367. [PMID: 39141663 DOI: 10.1080/1354750x.2024.2393342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is an inflammatory and necrotizing intestinal emergency that occurs in preterm infants and low birth weight newborns; however, no specific serum biomarkers for the diagnosis of NEC has been identified so far. METHODS Serum samples were collected from healthy neonatal controls and patients with NEC newly admitted to the Children's Hospital of Chongqing Medical University. ELISA was used to measure serum PK2 levels, and ROC curve analysis was sued to evaluate the diagnostic efficacy of PK2 and other clinical biomarkers. RESULTS Serum PK2 levels in the NEC group (n = 53) were significantly lower than those in the control group (n = 18), but increased to near-normal levels after the postoperative recovery period. The NLR value of NEC group was higher than that of control group (P < 0.05). There was no significant difference in WBC and PLT count between NEC group and control group (P > 0.05). Serum CRP and PCT levels in NEC group were significantly higher than those in control group (P < 0.001 for CRP and P < 0.05 for PCT, respectively). After surgery, serum CRP, NLR and PCT levels were lower than before surgery, while serum PK2 levels were higher than before surgery (P < 0.05). The areas under the ROC curve (AUC) of PK2, PCT and CRP for the diagnosis of NEC were 0.837, 0.662 and 0.552, respectively. The AUC of PK2 combined with PCT, PK2 combined with CRP, and PK2 combined with PCT and CRP were 0.908, 0.854 and 0.981, respectively. PK2 exhibited the highest diagnostic efficacy for NEC. CONCLUSION PK2 has higher diagnostic efficacy than PCT and CRP in the diagnosis of NEC; the combination of PK2 and PCT or CRP can significantly improve its diagnostic efficiency, especially when the three are combined at the same time.
Collapse
Affiliation(s)
- Qiuli Zeng
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Li Zeng
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Xiaoyan Yu
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Xi Yuan
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Wenjing Ma
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Zhixin Song
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Dapeng Chen
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| |
Collapse
|
31
|
Campos Martinez AM, Fernández Marín CE, Ruiz López A, Cubero Millán I, Uberos Fernández J. Evidence on the benefits of probiotics for preterm infants. NUTR HOSP 2024; 41:889-896. [PMID: 38967287 DOI: 10.20960/nh.03743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
Introduction This article reviews the evidence for the use of different strains of probiotics in the prevention of prevalent pathologies in premature neonates. A systematic review was conducted of the use of probiotics in neonates with less than 37 weeks of gestational age, based on a search for systematic reviews and observational and experimental studies performed during the period from January 2014 to February 2021. For this purpose, the PubMed, MEDLINE and Cochrane Library databases were consulted. The aim of this article was to review the existing data on the relationship between the administration of probiotics (with different strains and doses) and the risk of necrotising enterocolitis, mortality, late sepsis and other disease parameters in premature infants. The literature search obtained 240 articles, of which we selected 16, representing a total sample of over 200,000 premature infants. Analysis of the data obtained reveals statistical evidence that the combined administration of probiotics (especially of Lactobacillus and Bifidobacterium strains) reduces the incidence of grade II or higher necrotising enterocolitis, all-cause mortality, late sepsis, length of hospital stay and time until complete enteral nutrition is achieved. However, no benefits were apparent with respect to alleviating bronchopulmonary dysplasia, retinopathy of prematurity or intraventricular haemorrhage. Further research is needed to determine the most appropriate strains, doses and treatment duration for preterm infants to achieve the health benefits identified.
Collapse
Affiliation(s)
| | | | - Aida Ruiz López
- Neonatal Unit. Department of Pediatrics. Hospital Universitario Clínico San Cecilio
| | - Isabel Cubero Millán
- Neonatal Unit. Department of Pediatrics. Hospital Universitario Clínico San Cecilio
| | - José Uberos Fernández
- Neonatal Unit. Department of Pediatrics. Hospital Universitario Clínico San Cecilio. School of Medicine. Universidad de Granada
| |
Collapse
|
32
|
Srinivasjois R, Gebremedhin A, Silva D, Rao SC, Tessema GA, Pereira G. Probiotic Supplementation in the Neonatal Age Group and the Risk of Hospitalisation in the First Two Years: A Data Linkage Study from Western Australia. Nutrients 2024; 16:2094. [PMID: 38999842 PMCID: PMC11243646 DOI: 10.3390/nu16132094] [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: 05/30/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Probiotic supplementation in preterm neonates is standard practice in many centres across the globe. The impact of probiotic supplementation in the neonatal age group on the risk of hospitalisation in infancy has not been reported previously. METHODS Infants born < 32 + 6 weeks of gestation in Western Australia were eligible for inclusion. We conducted a retrospective cohort study comparing data from before probiotic supplementation (Epoch 1: 1 December 2008-30 November 2010, n = 1238) versus after (Epoch 2: 1 June 2012-30 May 2014, n = 1422) on the risks of respiratory- and gastrointestinal infection-related hospitalisation. A subgroup analysis of infants born < 28 weeks of gestation was analysed separately for similar outcomes. RESULTS Compared to Epoch 1, an 8% reduction in incidence of hospitalisation up to 2 years after birth was observed in Epoch 2 (adjusted incidence rate ratio (IRR) of 0.92; 95% confidence interval (CI); 0.87-0.98), adjusted for gestational age, smoking, socioeconomic status, and maternal age. The rate of hospitalisation for infants born < 28 weeks of gestation was comparable in epochs 1 and 2. CONCLUSION Infants exposed to probiotic supplementation in the neonatal period experience a reduced risk of hospitalisation in the first two years after discharge from the neonatal unit.
Collapse
Affiliation(s)
- Ravisha Srinivasjois
- Curtin School of Population Health, Curtin University, Perth, WA 6102, Australia; (A.G.); (G.A.T.); (G.P.)
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Perth, WA 6027, Australia;
- School of Medicine, University of Western Australia, Perth, WA 6005, Australia;
| | - Amanuel Gebremedhin
- Curtin School of Population Health, Curtin University, Perth, WA 6102, Australia; (A.G.); (G.A.T.); (G.P.)
- EnAble Institute, Curtin University, Perth, WA 6102, Australia
| | - Desiree Silva
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Perth, WA 6027, Australia;
- School of Medicine, University of Western Australia, Perth, WA 6005, Australia;
| | - Shripada C. Rao
- School of Medicine, University of Western Australia, Perth, WA 6005, Australia;
- Neonatal Directorate, Child and Adolescent Health Service, Perth Children’s Hospital, Perth, WA 6009, Australia
| | - Gizachew A. Tessema
- Curtin School of Population Health, Curtin University, Perth, WA 6102, Australia; (A.G.); (G.A.T.); (G.P.)
- EnAble Institute, Curtin University, Perth, WA 6102, Australia
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Perth, WA 6102, Australia; (A.G.); (G.A.T.); (G.P.)
- EnAble Institute, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
33
|
Chen W, Guo K, Huang X, Zhang X, Li X, Chen Z, Wang Y, Wang Z, Liu R, Qiu H, Wang M, Zeng S. The Association of Neonatal Gut Microbiota Community State Types with Birth Weight. CHILDREN (BASEL, SWITZERLAND) 2024; 11:770. [PMID: 39062221 PMCID: PMC11276374 DOI: 10.3390/children11070770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND while most gut microbiota research has focused on term infants, the health outcomes of preterm infants are equally important. Very-low-birth-weight (VLBW) or extremely-low-birth-weight (ELBW) preterm infants have a unique gut microbiota structure, and probiotics have been reported to somewhat accelerate the maturation of the gut microbiota and reduce intestinal inflammation in very-low preterm infants, thereby improving their long-term outcomes. The aim of this study was to investigate the structure of gut microbiota in ELBW neonates to facilitate the early identification of different types of low-birth-weight (LBW) preterm infants. METHODS a total of 98 fecal samples from 39 low-birth-weight preterm infants were included in this study. Three groups were categorized according to different birth weights: ELBW (n = 39), VLBW (n = 39), and LBW (n = 20). The gut microbiota structure of neonates was obtained by 16S rRNA gene sequencing, and microbiome analysis was conducted. The community state type (CST) of the microbiota was predicted, and correlation analysis was conducted with clinical indicators. Differences in the gut microbiota composition among ELBW, VLBW, and LBW were compared. The value of gut microbiota composition in the diagnosis of extremely low birth weight was assessed via a random forest-machine learning approach. RESULTS we briefly analyzed the structure of the gut microbiota of preterm infants with low birth weight and found that the ELBW, VLBW, and LBW groups exhibited gut microbiota with heterogeneous compositions. Low-birth-weight preterm infants showed five CSTs dominated by Enterococcus, Staphylococcus, Klebsiella, Streptococcus, Pseudescherichia, and Acinetobacter. The birth weight and clinical indicators related to prematurity were associated with the CST. We found the composition of the gut microbiota was specific to the different types of low-birth-weight premature infants, namely, ELBW, VLBW, and LBW. The ELBW group exhibited significantly more of the potentially harmful intestinal bacteria Acinetobacter relative to the VLBW and LBW groups, as well as a significantly lower abundance of the intestinal probiotic Bifidobacterium. Based on the gut microbiota's composition and its correlation with low weight, we constructed random forest model classifiers to distinguish ELBW and VLBW/LBW infants. The area under the curve of the classifiers constructed with Enterococcus, Klebsiella, and Acinetobacter was found to reach 0.836 by machine learning evaluation, suggesting that gut microbiota composition may be a potential biomarker for ELBW preterm infants. CONCLUSIONS the gut bacteria of preterm infants showed a CST with Enterococcus, Klebsiella, and Acinetobacter as the dominant genera. ELBW preterm infants exhibit an increase in the abundance of potentially harmful bacteria in the gut and a decrease in beneficial bacteria. These potentially harmful bacteria may be potential biomarkers for ELBW preterm infants.
Collapse
Affiliation(s)
- Wanling Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518116, China
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
| | - Kaiping Guo
- Division of Pediatrics, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xueli Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Xiaoxia Li
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Zimiao Chen
- Department of Burn Plastic Surgery, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Yanli Wang
- Department of Pediatrics, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Rongtian Liu
- Department of Pediatrics, Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
- Department of Neonatology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen 518172, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| |
Collapse
|
34
|
TAKESHITA K, TAKEI H, TANAKA S, HISHIKI H, IIJIMA Y, OGATA H, FUJISHIRO K, TOMINAGA T, KONNO Y, IWASE Y, ENDO M, ISHIWADA N, OSONE Y, TAKEMURA R, HAMADA H, SHIMOJO N. Effect of multi-strain bifidobacteria supplementation on intestinal microbiota development in low birth weight neonates: a randomized controlled trial. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:352-358. [PMID: 39364130 PMCID: PMC11444860 DOI: 10.12938/bmfh.2023-093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/21/2024] [Indexed: 10/05/2024]
Abstract
Single-strain Bifidobacterium species are commonly used as probiotics with low birth weight neonates. However, the effectiveness and safety of multi-strain Bifidobacterium supplementation are not well known. Thirty-six neonates weighing less than 2,000 g (558-1,943 g) at birth and admitted to a neonatal intensive care unit were randomly assigned to receive a single strain or triple strains of Bifidobacterium with lactulose enterally for 4 weeks from birth. The relative abundances of Staphylococcus and Bifidobacterium in the fecal microbiota at weeks 1, 2, and 4 were investigated. Based on the study results, no significant difference was detected between the two groups in the abundance of Staphylococcus; however, the triple-strain group had significantly high abundances of Bifidobacterium at weeks 2 and 4. The fecal microbiota in the triple-strain group had significantly lower alpha diversity (Bifidobacterium-enriching) after week 4 and was different from that in the single-strain group, which showed a higher abundance of Clostridium. No severe adverse events occurred in either group during the study period. Although no significant difference was detected between single- and multi-strain bifidobacteria supplementation in the colonization of Staphylococcus in the fecal microbiota of the neonates, multi-strain bifidobacteria supplementation contributed toward early enrichment of the microbiota with bifidobacteria and suppression of other pathogenic bacteria, such as Clostridium spp.
Collapse
Affiliation(s)
- Kenichi TAKESHITA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Department of Pediatrics, Chiba Rosai Hospital, 2-16
Tatsumidaihigashi, Ichihara-shi, Chiba 290-0003, Japan
| | - Haruka TAKEI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Saori TANAKA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka HISHIKI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Yuta IIJIMA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Hitoshi OGATA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Kensuke FUJISHIRO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Takahiro TOMINAGA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yuki KONNO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yukiko IWASE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Mamiko ENDO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Naruhiko ISHIWADA
- Department of Infectious Diseases, Medical Mycology Research
Center, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8673, Japan
| | - Yoshiteru OSONE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Ryo TAKEMURA
- Clinical and Translational Research Center, Keio University
Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiromichi HAMADA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Naoki SHIMOJO
- Center for Preventive Medical Sciences, Chiba University,
1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| |
Collapse
|
35
|
Sánchez-González SG, Cárdenas-del-Castillo BG, Garza-González E, Padilla-Rivas GR, Rodríguez-Balderrama I, Treviño-Garza C, Montes-Tapia FF, Palacios-Saucedo GC, Gutiérrez-Rodríguez A, de-la-O-Cavazos ME. Gut microbiota in preterm infants receiving breast milk or mixed feeding. World J Clin Pediatr 2024; 13:90499. [PMID: 38947995 PMCID: PMC11212766 DOI: 10.5409/wjcp.v13.i2.90499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Preterm birth is the leading cause of mortality in newborns, with very-low-birth-weight infants usually experiencing several complications. Breast milk is considered the gold standard of nutrition, especially for preterm infants with delayed gut colonization, because it contains beneficial microorganisms, such as Lactobacilli and Bifidobacteria. AIM To analyze the gut microbiota of breastfed preterm infants with a birth weight of 1500 g or less. METHODS An observational study was performed on preterm infants with up to 36.6 wk of gestation and a birth weight of 1500 g or less, born at the University Hospital Dr. José Eleuterio González at Monterrey, Mexico. A total of 40 preterm neonates were classified into breast milk feeding (BM) and mixed feeding (MF) groups (21 in the BM group and 19 in the MF group), from October 2017 to June 2019. Fecal samples were collected before they were introduced to any feeding type. After full enteral feeding was achieved, the composition of the gut microbiota was analyzed using 16S rRNA gene sequencing. Numerical variables were compared using Student's t-test or using the Mann-Whitney U test for nonparametric variables. Dominance, evenness, equitability, Margalef's index, Fisher's alpha, Chao-1 index, and Shannon's diversity index were also calculated. RESULTS No significant differences were observed at the genus level between the groups. Class comparison indicated higher counts of Alphaproteobacteria and Betaproteobacteria in the initial compared to the final sample of the BM group (P < 0.011). In addition, higher counts of Gammaproteobacteria were detected in the final than in the initial sample (P = 0.040). According to the Margalef index, Fisher's alpha, and Chao-1 index, a decrease in species richness from the initial to the final sample, regardless of the feeding type, was observed (P < 0.050). The four predominant phyla were Bacteroidetes, Actinobacteria, Firmicutes, and Proteobacteria, with Proteobacteria being the most abundant. However, no significant differences were observed between the initial and final samples at the phylum level. CONCLUSION Breastfeeding is associated with a decrease in Alphaproteobacteria and Betaproteobacteria and an increase of Gammaproteobacteria, contributing to the literature of the gut microbiota structure of very low-birth-weight, preterm.
Collapse
Affiliation(s)
- Sandra Gabriela Sánchez-González
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | | | - Elvira Garza-González
- Departamento de Bioquímica y Medicina Molecular, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Gerardo R Padilla-Rivas
- Department of Biochemistry and Molecular Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Isaías Rodríguez-Balderrama
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Consuelo Treviño-Garza
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Fernando Félix Montes-Tapia
- Department of Pediatric Surgery, Hospital Universitario "Dr. José Eleuterio González", Monterrey 64460, Nuevo León, México
| | - Gerardo C Palacios-Saucedo
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Anthony Gutiérrez-Rodríguez
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Manuel Enrique de-la-O-Cavazos
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| |
Collapse
|
36
|
He P, Yu L, Tian F, Chen W, Zhang H, Zhai Q. Effects of Probiotics on Preterm Infant Gut Microbiota Across Populations: A Systematic Review and Meta-Analysis. Adv Nutr 2024; 15:100233. [PMID: 38908894 PMCID: PMC11251410 DOI: 10.1016/j.advnut.2024.100233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 06/24/2024] Open
Abstract
Microbiota in early life is closely associated with the health of infants, especially premature ones. Probiotics are important drivers of gut microbiota development in preterm infants; however, there is no consensus regarding the characteristics of specific microbiota in preterm infants receiving probiotics. In this study, we performed a meta-analysis of 5 microbiome data sets (1816 stool samples from 706 preterm infants) to compare the gut microbiota of preterm infants exposed to probiotics with that of preterm infants not exposed to probiotics across populations. Despite study-specific variations, we found consistent differences in gut microbial composition and predicted functional pathways between the control and probiotic groups across different cohorts of preterm infants. The enrichment of Acinetobacter, Bifidobacterium, and Lactobacillus spp and the depletion of the potentially pathogenic bacteria Finegoldia, Veillonella, and Klebsiella spp. were the most consistent changes in the gut microbiota of preterm infants supplemented with probiotics. Probiotics drove microbiome transition into multiple preterm gut community types, and notably, preterm gut community type 3 had the highest α-diversity, with enrichment of Bifidobacterium and Bacteroides spp. At the functional level, the major predicted microbial pathways involved in peptidoglycan biosynthesis consistently increased in preterm infants supplemented with probiotics; in contrast, the crucial pathways associated with heme biosynthesis consistently decreased. Interestingly, Bifidobacterium sp. rather than Lactobacillus sp. gradually became dominant in gut microbiota of preterm infants using mixed probiotics, although both probiotic strains were administered at the same dosage. Taken together, our meta-analysis suggests that probiotics contribute to reshaping the microbial ecosystem of preterm infants at both the taxonomic and functional levels of the bacterial community. More standardized and relevant studies may contribute to better understanding the crosstalk among probiotics, the gut microbiota, and subsequent disease risk, which could help to give timely nutritional feeding guidance to preterm infants. This systematic review and meta-analysis was registered at PROSPERO (https://www.crd.york.ac.uk/PROSPERO/) as CRD42023447901.
Collapse
Affiliation(s)
- Pandi He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China.
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
37
|
van den Akker CHP, Embleton ND, Lapillonne A, Mihatsch WA, Salvatore S, Canani RB, Dinleyici EC, Domellöf M, Guarino A, Gutiérrez-Castrellón P, Hojsak I, Indrio F, Mosca A, Orel R, van Goudoever JHB, Weizman Z, Mader S, Zimmermann LJI, Shamir R, Vandenplas Y, Szajewska H. Reevaluating the FDA's warning against the use of probiotics in preterm neonates: A societal statement by ESPGHAN and EFCNI. J Pediatr Gastroenterol Nutr 2024; 78:1403-1408. [PMID: 38572770 DOI: 10.1002/jpn3.12204] [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: 01/02/2024] [Revised: 02/14/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024]
Abstract
The recent advisory issued by the United States Food and Drug Administration, cautioning against the routine administration of probiotics in preterm neonates, has sparked a lively debate within the scientific community. This commentary presents a perspective from members of the Special Interest Group on Gut Microbiota and Modifications within the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) and other authors who contributed to the ESPGHAN position paper on probiotics for preterm infants, as well as representatives from the European Foundation for the Care of Newborn Infants. We advocate for a more nuanced and supportive approach to the use of certain probiotics in this vulnerable population, balancing the demonstrated benefits and risks.
Collapse
Affiliation(s)
- Chris H P van den Akker
- Department of Pediatrics-Neonatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, and Amsterdam Gastroenterology Endocrinology Metabolism Research Institutes, Amsterdam, The Netherlands
| | - Nicholas D Embleton
- Neonatal Unit, Royal Victoria Infirmary, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Alexandre Lapillonne
- Department of Neonatology, APHP Necker University Hospital, Paris Cite University, Paris, France
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Walter A Mihatsch
- Department of Pediatrics, Ulm University, Ulm, Germany
- Department of Health Management, Neu-Ulm University of Applied Sciences, Neu-Ulm, Germany
- Department of Pediatrics, Zollernalb Klinikum, Balingen, Germany
| | - Silva Salvatore
- Department of Medicine and Technological Innovation, Pediatrics, Hospital "F. Del Ponte", University of Insubria, Varese, Italy
| | - Roberto B Canani
- Department of Translational Medical Science and the ImmunoNutritionLab at the Ceinge Research Center and Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Ener C Dinleyici
- Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Alfredo Guarino
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | | | - Iva Hojsak
- Department of Pediatrics, University of Zagreb Medical School, Children's Hospital Zagreb, Zagreb, Croatia
| | - Flavia Indrio
- Department of Experimental Medicine, Pediatric Section, Medical School MedTech, University of Salento, Lecce, Italy
| | - Alexis Mosca
- Department Pediatric Gastroenterology and Nutrition, Robert-Debré Hospital, APHP, Paris, France
| | - Rok Orel
- Department of Gastroenterology, Hepatology and Nutrition, University Medical Center Ljubljana, University Children's Hospital Ljubljana, Medical Faculty, University of Ljubljana, Ljubeljana, Slovenia
| | - Johannes Hans B van Goudoever
- Department of Pediatrics, Amsterdam UMC, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Zvi Weizman
- Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Silke Mader
- European Foundation for the Care of Newborn Infants (EFCNI), Munich, Germany
| | - Luc J I Zimmermann
- European Foundation for the Care of Newborn Infants (EFCNI), Munich, Germany
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht UMC+, Maastricht, The Netherlands
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yvan Vandenplas
- Department of Pediatrics, UZ Brussel, KidZ Health Castle, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hania Szajewska
- Department of Pediatrics, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
38
|
Lee CC, Chiu CH. Link between gut microbiota and neonatal sepsis. J Formos Med Assoc 2024; 123:638-646. [PMID: 37821302 DOI: 10.1016/j.jfma.2023.09.019] [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: 05/26/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
In neonates, the gastrointestinal tract is rapidly colonized by bacteria after birth. Gut microbiota development is critical during the first few years of life. However, disruption of gut microbiota development in neonates can lead to gut dysbiosis, characterized by overcolonization by pathogenic bacteria and delayed or failed maturation toward increasing microbial diversity and Fermicutes dominance. Gut dysbiosis can predispose infants to sepsis. Pathogenic bacteria can colonize the gut prior to sepsis and cause sepsis through translocation. This review explores gut microbiota development in neonates, the evidence linking gut dysbiosis to neonatal sepsis, and the potential role of probiotics in gut microbiota modulation and sepsis prevention.
Collapse
Affiliation(s)
- Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
39
|
Tanaka S, Takahashi M, Takeshita K, Nagasawa K, Takei H, Sato H, Hishiki H, Ishiwada N, Hamada H, Kadota Y, Tochio T, Ishida T, Sasaki K, Tomita M, Osone Y, Takemura R, Shimojo N. The prebiotic effect of 1-kestose in low-birth-weight neonates taking bifidobacteria: a pilot randomized trial in comparison with lactulose. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:329-335. [PMID: 39364124 PMCID: PMC11444857 DOI: 10.12938/bmfh.2023-079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/29/2024] [Indexed: 10/05/2024]
Abstract
Probiotics such as bifidobacteria have been given to low-birth-weight neonates (LBWNs) at risk for a disrupted gut microbiota leading to the development of serious diseases such necrotizing enterocolitis. Recently prebiotics such as lactulose are used together with bifidobacteria as synbiotics. However, faster and more powerful bifidobacteria growth is desired for better LBWN outcomes. The prebiotic 1-kestose has a higher selective growth-promoting effect on bifidobacteria and lactic acid bacteria in vitro among several oligosaccharides. Twenty-six premature neonates (less than 2,000 g) admitted to a neonatal intensive care unit (NICU) were randomly assigned to receive Bifidobacterium breve M16-V with either 1-kestose or lactulose once a day for four weeks from birth. A 16S rRNA gene analysis revealed similar increases in alpha-diversity from 7 to 28 days in both groups. The most dominant genus on both days was Bifidobacterium in both groups, with no significant difference between the two groups. Quantitative PCR analysis revealed that the number of Staphylococcus aureus tended to be lower in the 1-kestose group than in the lactulose group at 28 days. The number of Escherichia coli was higher in the 1-kestose group at 7 days. The copy number of total bacteria in the 1-kestose group was significantly higher than that in the lactulose group at 3 time points, 7, 14, and 28 days. No severe adverse events occurred in either group during the study period. l-Ketose may offer an alternative option to lactulose as a prebiotic to promote the development of gut microbiota in LBWNs.
Collapse
Affiliation(s)
- Saori Tanaka
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Mayuko Takahashi
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Kenichi Takeshita
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Koo Nagasawa
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka Takei
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Hironori Sato
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka Hishiki
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Naruhiko Ishiwada
- Department of Infectious Diseases, Medical Mycology Research Center, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8673, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Yoshihiro Kadota
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Takumi Tochio
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Tomoki Ishida
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Koh Sasaki
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Mika Tomita
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Yoshiteru Osone
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Ryo Takemura
- Clinical and Translational Research Center, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Naoki Shimojo
- Center for Preventive Medical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| |
Collapse
|
40
|
Govindaraj D, Jensen GB, Rahman Qazi K, Sverremark‐Ekström E, Abrahamsson T, Jenmalm MC. Effects of extremely preterm birth on cytokine and chemokine responses induced by T-cell activation during infancy. Clin Transl Immunology 2024; 13:e1510. [PMID: 38737447 PMCID: PMC11087183 DOI: 10.1002/cti2.1510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
Objectives Extremely preterm (EPT; gestational week < 28 + 0, < 1000 g) neonates are vulnerable to infections and necrotising enterocolitis, important contributors to mortality and morbidity. However, knowledge regarding their immune maturation remains limited. We here investigated the longitudinal development of functional T-cell capacity in EPT infants. Methods Peripheral blood mononuclear cells were isolated at 14th and 28th day (D) and at gestational week 36 + 0 (Gw36) from EPT infants, participated in a randomised, double-blind, placebo-controlled study of Lactobacillus reuteri DSM 17938 probiotic supplementation. Blood collected from 25 full-term (FT) infants at D14 was used as control. The secretion of immune mediators was determined through comprehensive Luminex panels after stimulation with human T-cell activator CD3/CD28 beads. Results The levels of many mediators were low in EPT infants at D14, whereas the secretion of several chemokines was higher in EPT than in FT infants. Furthermore, Th2:Th1 cytokine ratios were higher in EPT than in FT infants. Progressively elevated secretion of, for example, IFN-γ, TNF and IL-17A in EPT infants was observed from D14 to D28 and then at Gw36. Elevated levels were observed for many proinflammatory mediators at D28. Probiotic supplementation or perinatal factors (e.g. clinical chorioamnionitis, preeclampsia and delivery mode) did not influence the cytokine and chemokine responses. Conclusions Immune mediators induced by T-cell activation in EPT infants were mainly reduced at D14 and Th2 skewed compared to those in FT infants, but mostly recovered at Gw36, indicating immune maturation. Increased proinflammatory responses at D28 may be related to the heightened risk of severe immune-associated complications seen in EPT infants.
Collapse
Affiliation(s)
- Dhanapal Govindaraj
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| | - Georg Bach Jensen
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Eva Sverremark‐Ekström
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Thomas Abrahamsson
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| |
Collapse
|
41
|
Wu WH, Lee CC, Chen YC, Chiang MC, Chiu CH. Invasive lactobacillus infection in pediatric patients in a tertiary center in Taiwan - 16 years' experience and literature review. Pediatr Neonatol 2024; 65:282-287. [PMID: 38007356 DOI: 10.1016/j.pedneo.2023.05.013] [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: 03/22/2023] [Revised: 05/06/2023] [Accepted: 05/19/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Lactobacilli are common microorganisms in the human body. Some species were used as probiotics supplement for many purposes such as preventing necrotizing enterocolitis, or improving allergic diseases or diarrhea. Previously, Lactobacillus infection was thought of as contamination due to its low pathogenicity. However, there have been reports of invasive Lactobacillus infection in immunocompromised patients or patients with comorbidities. The purpose of this study was to analyze the clinical characteristics, antibiotic treatment and outcomes of pediatric patients with invasive Lactobacillus infection. METHODS We retrospectively reviewed pediatric patients diagnosed with invasive Lactobacillus infection between 2004 and 2020. Invasive Lactobacillus infection was diagnosed if sterile sites yielded Lactobacillus spp. Clinical manifestations, chronic diseases, potential predisposing factors, medical treatments, antimicrobial susceptibility tests and outcomes were recorded. RESULTS Fifteen pediatric patients were diagnosed with invasive Lactobacillus infection, accounting for 2.4% of total invasive Lactobacillus infections during the 16-year period. Eleven infections were bacteremia, two were intra-abdominal infections, and two were biliary tract infections. Fever was the most common symptom. Potential predisposing factors were immunocompromised status, central venous device, prolonged antibiotics use and receiving supplemented probiotics for at least one week. All patients survived with favorable outcomes. Most pathogens were identified as Lactobacillus spp, and two were Lactobacillus rhamnosus, which were related to supplemented probiotics. The antimicrobial susceptibility tests showed that Lactobacilli were all sensitive to ampicillin but resistant to glycopeptides. CONCLUSION Invasive Lactobacillus infections in pediatric patients were rare. Despite its low pathogenicity, Lactobacillus could cause invasive infection in those immunocompromised patients.
Collapse
Affiliation(s)
- Wei-Hung Wu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Ching Chen
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan; Division of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
42
|
Neu J, Ashley P, Chowdhary V, Lampland A, Porcelli P, Rothstein R, Slancheva B, Kronström A, Rastad J, Strömberg S, Thuresson M. Progression of Enteral Feeding Volumes in Extremely Low Birth Weight Infants in the "Connection Trial". Am J Perinatol 2024; 41:e2717-e2726. [PMID: 37683670 PMCID: PMC11663079 DOI: 10.1055/s-0043-1774314] [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: 03/03/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
OBJECTIVE Investigate daily feeding volumes and their association with clinical variables in the early postnatal care of premature infants of the "Connection Trial." STUDY DESIGN A total of 641 infants of 510 to 1,000-g birth weight (BW, mean: 847 g) and mean 27 weeks' gestational age at birth (GA) were analyzed for total daily enteral (TDE) feeding volumes of 10, 20, 40, 80, and 120 mL/kg/d and their association with 24 clinical variables. Uni- and multivariable Cox regression models were used to calculate hazard ratios (HR) with 95% confidence intervals as a measure of the chance of reaching each of the TDE volumes. RESULTS Daily feeding volumes were highly variable and the median advancement from 10 to 120 mL/kg/d was 11 mL/kg/d. Univariable analyses showed the lowest chance (HR, 0.22-0.81) of reaching the TDE volumes for gastrointestinal (GI) serious adverse events (SAEs), GI perforation, GI obstruction, and necrotizing enterocolitis, as well as respiratory SAEs, persistent ductus arteriosus, and hypotension. Each GA week, 100-g BW, and point in 5-minute Apgar score at birth associated with 8 to 20% increased chance of reaching the TDE volumes. Multivariable analyses showed independent effects for BW, GA, Apgar score, GI SAEs, abdominal symptoms/signs, respiratory SAEs, days on antibiotics, and hypotension. CONCLUSION This observational analysis demonstrates the variable and cautious progression of enteral feedings in contemporary extremely low BW infants and the extent to which clinical variables associate with this progression. KEY POINTS · Total feedings of 10 and 120 mL/kg/d were reached at median 4 and 14 day of age, respectively, and at a daily increase of 11 mL/kg.. · Each incremental GA week, 100-g BW, and point in 5-minute Apgar score associated with 8 to 20% increased chance of reaching enteral feedings of 10 to 120 mL/kg/d.. · Progression of enteral feeding associated with several clinical events and was slower than advocated in common feeding protocols..
Collapse
Affiliation(s)
- Josef Neu
- Department of Pediatrics, University of Florida Health Shands Children's Hospital, Gainesville, Florida
| | - Patricia Ashley
- Department of Pediatrics, Duke University, Durham, North Carolina
| | - Vikas Chowdhary
- Department of Pediatrics, Arkansas Children's Hospital, Little Rock, Arkansas
| | - Andrea Lampland
- Department of Neonatology, Children's Minnesota St. Paul Clinic, Saint Paul, Minnesota
| | - Peter Porcelli
- Department of Pediatrics, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, North Carolina
| | - Robert Rothstein
- Department of Pediatrics, Baystate Children's Hospital, Springfield, Massachusetts
| | - Boriana Slancheva
- Department of Neonatology, Medical University of Sofia, Sofia, Bulgaria
| | | | - Jonas Rastad
- Infant Bacterial Therapeutics Inc., Stockholm, Sweden
| | | | | | | |
Collapse
|
43
|
Roberts AG, Younge N, Greenberg RG. Neonatal Necrotizing Enterocolitis: An Update on Pathophysiology, Treatment, and Prevention. Paediatr Drugs 2024; 26:259-275. [PMID: 38564081 DOI: 10.1007/s40272-024-00626-w] [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] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease predominantly affecting premature and very low birth weight infants resulting in inflammation and necrosis of the small bowel and colon and potentially leading to sepsis, peritonitis, perforation, and death. Numerous research efforts have been made to better understand, treat, and prevent NEC. This review explores a variety of factors involved in the pathogenesis of NEC (prematurity, low birth weight, lack of human breast milk exposure, alterations to the microbiota, maternal and environmental factors, and intestinal ischemia) and reports treatment modalities surrounding NEC, including pain medications and common antibiotic combinations, the rationale for these combinations, and recent antibiotic stewardship approaches surrounding NEC treatment. This review also highlights the effect of early antibiotic exposure, infections, proton pump inhibitors (PPIs), and H2 receptor antagonists on the microbiota and how these risk factors can increase the chances of NEC. Finally, modern prevention strategies including the use of human breast milk and standardized feeding regimens are discussed, as well as promising new preventative and treatment options for NEC including probiotics and stem cell therapy.
Collapse
|
44
|
Hanna M, Ahmad I, Yanowitz T, Kim J, Hunter C, DiGeronimo R, Ahmad KA, Sullivan K, Markel TA, Hair AB, Chaaban H, Pammi M, Huff KA, Jasani B, Fuchs L, Cuna A, Garg PM, Reber K, Premkumar MH. Current Patterns of Probiotic Use in U.S. Neonatal Intensive Care Units: A Multi-Institution Survey. Am J Perinatol 2024; 41:e2658-e2665. [PMID: 37494969 DOI: 10.1055/a-2140-8727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
OBJECTIVE Probiotic supplementation is associated with health benefits in preterm infants. The 2021 American Academy of Pediatrics (AAP) statement on probiotic use advised caution, citing heterogeneity and absence of federal regulation. We assessed the impact of the AAP statement and current institution-wide patterns of probiotic use across neonatal intensive care units (NICU) across the United States. STUDY DESIGN A cross-sectional web-based institutional survey using REDCap was emailed to 430 Children's Hospital Neonatal Consortium (CHNC) and Pediatrix Medical Group institutions. The survey captured data on probiotic formulations, supplementation, initiation and cessation criteria, reasons for discontinuation, interest in initiating, and AAP statement's impact. RESULTS Ninety-five (22.1%) hospitals, including 42/46 (91%) CHNC and 53/384 (14%) Pediatrix institutions, completed the survey. Thirty-seven (39%) currently use probiotics. Fourteen different probiotic formulations were reported. The common criteria for initiation were birth weight <1,500 g and gestational age <32 weeks. Parental consent or assent was obtained at only 30% of institutions. Five hospitals (11%) with prior probiotic use discontinued solely due to the AAP statement. Overall, 23 (24%) of hospitals indicated that the AAP statement significantly influenced their decision regarding probiotic use. Nineteen of 51 nonusers (37%) are considering initiation. CONCLUSION Probiotic use in preterm infants is likely increasing in NICUs across the United States, but significant variability exists. The 2021 AAP statement had variable impact on NICUs' decision regarding probiotic use. The growing interest in adopting probiotics and the significant interhospital variability highlight the need for better regulation and consensus guidelines to ensure standardized use. KEY POINTS · Probiotic use in preterm infants is likely increasing in U.S. NICUs, but clinical variability exists.. · The AAP statement on probiotic use in preterm infants had a modest impact on current practices.. · There's a need for better product regulation and consensus guidelines to ensure standardized use..
Collapse
Affiliation(s)
- Morcos Hanna
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Irfan Ahmad
- Division of Neonatology, Children's Hospitals Orange County, Orange, California
| | - Toby Yanowitz
- Division of Neonatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jae Kim
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catherine Hunter
- Division of Pediatric Surgery, Department of Surgery, Oklahoma Children's Hospital, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Robert DiGeronimo
- Division of Neonatology, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, Washington
| | - Kaashif A Ahmad
- Department of Pediatrics, Pediatrix and Obstetrix Specialists of Houston, Houston, Texas
| | - Kevin Sullivan
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
- Division of Neonatology, Nemours/AI duPont Hospital for Children, Wilmington, Delaware
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Amy B Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Hala Chaaban
- Division of Neonatology, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mohan Pammi
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Katie A Huff
- Division of Neonatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bonny Jasani
- Division of Neonatology, Hospital for Sick Children, Toronto, Canada
| | - Lynn Fuchs
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
- Division of Neonatology, Nemours/AI duPont Hospital for Children, Wilmington, Delaware
| | - Alain Cuna
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, Missouri
| | - Parvesh M Garg
- Division of Neonatology, Wake Forest University, Winston Salem, North Carolina
| | - Kristina Reber
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Muralidhar H Premkumar
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
45
|
Sizemore N, Oliphant K, Zheng R, Martin CR, Claud EC, Chattopadhyay I. A digital twin of the infant microbiome to predict neurodevelopmental deficits. SCIENCE ADVANCES 2024; 10:eadj0400. [PMID: 38598636 PMCID: PMC11006218 DOI: 10.1126/sciadv.adj0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
Despite the recognized gut-brain axis link, natural variations in microbial profiles between patients hinder definition of normal abundance ranges, confounding the impact of dysbiosis on infant neurodevelopment. We infer a digital twin of the infant microbiome, forecasting ecosystem trajectories from a few initial observations. Using 16S ribosomal RNA profiles from 88 preterm infants (398 fecal samples and 32,942 abundance estimates for 91 microbial classes), the model (Q-net) predicts abundance dynamics with R2 = 0.69. Contrasting the fit to Q-nets of typical versus suboptimal development, we can reliably estimate individual deficit risk (Mδ) and identify infants achieving poor future head circumference growth with ≈76% area under the receiver operator characteristic curve, 95% ± 1.8% positive predictive value at 98% specificity at 30 weeks postmenstrual age. We find that early transplantation might mitigate risk for ≈45.2% of the cohort, with potentially negative effects from incorrect supplementation. Q-nets are generative artificial intelligence models for ecosystem dynamics, with broad potential applications.
Collapse
Affiliation(s)
- Nicholas Sizemore
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Kaitlyn Oliphant
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Ruolin Zheng
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Camilia R. Martin
- Division of Neonatology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Erika C. Claud
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
- Neonatology Research, University of Chicago, Chicago, IL 60637, USA
| | - Ishanu Chattopadhyay
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Committee on Quantitative Methods in Social, Behavioral, and Health Sciences, University of Chicago, Chicago, IL 60637, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
- Center for Health Statistics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
46
|
Mackay CA, Rath C, Rao S, Patole S. Plant-Derived Substances for Prevention of Necrotising Enterocolitis: A Systematic Review of Animal Studies. Nutrients 2024; 16:832. [PMID: 38542743 PMCID: PMC10975714 DOI: 10.3390/nu16060832] [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: 01/29/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Inflammation, oxidative injury, and gut dysbiosis play an important role in the pathogenesis of necrotising enterocolitis (NEC). Plant-derived substances have historically been used as therapeutic agents due to their anti-inflammatory, antioxidant, and antimicrobial properties. We aimed to review pre-clinical evidence for plant-derived substances in the prevention and treatment of NEC. A systematic review was conducted using the following databases: PubMed, EMBASE, EMCARE, MEDLINE and Cochrane Library (PROSPERO CRD42022365477). Randomized controlled trials (RCTs) and quasi-RCTs that evaluated a plant-derived substance as an intervention for NEC in an animal model of the illness and compared pre-stated outcomes (e.g., clinical severity, severity of intestinal injury, mortality, laboratory markers of inflammation and oxidative injury) were included. Sixteen studies (n = 610) were included in the systematic review. Ten of the sixteen included RCTs (Preterm rat pups: 15, Mice: 1) reported mortality and all reported NEC-related histology. Meta-analysis showed decreased mortality [12/134 vs. 27/135; RR: 0.48 (95% CI: 0.26 to 0.87); p = 0.02, 10 RCTs] and decreased NEC in the experimental group [24/126 vs. 55/79; RR: 0.34 (95% CI: 0.22 to 0.52); p < 0.001, 6 RCTs]. Markers of inflammation (n = 11) and oxidative stress (n = 13) improved in all the studies that have reported this outcome. There was no significant publication bias for the outcome of mortality. Plant-derived substances have the potential to reduce the incidence and severity of histologically diagnosed NEC and mortality in rodent models. These findings are helpful in guiding further pre-clinical studies towards developing a food supplement for the prevention of NEC in preterm infants.
Collapse
Affiliation(s)
| | - Chandra Rath
- Neonatology, King Edward Memorial Hospita, Subiaco 6008, Australia
- Perth Children’s Hospital, Nedlands 6009, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| | - Shripada Rao
- Perth Children’s Hospital, Nedlands 6009, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| | - Sanjay Patole
- Neonatology, King Edward Memorial Hospita, Subiaco 6008, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| |
Collapse
|
47
|
Catassi G, Aloi M, Giorgio V, Gasbarrini A, Cammarota G, Ianiro G. The Role of Diet and Nutritional Interventions for the Infant Gut Microbiome. Nutrients 2024; 16:400. [PMID: 38337684 PMCID: PMC10857663 DOI: 10.3390/nu16030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The infant gut microbiome plays a key role in the healthy development of the human organism and appears to be influenced by dietary practices through multiple pathways. First, maternal diet during pregnancy and infant nutrition significantly influence the infant gut microbiota. Moreover, breastfeeding fosters the proliferation of beneficial bacteria, while formula feeding increases microbial diversity. The timing of introducing solid foods also influences gut microbiota composition. In preterm infants the gut microbiota development is influenced by multiple factors, including the time since birth and the intake of breast milk, and interventions such as probiotics and prebiotics supplementation show promising results in reducing morbidity and mortality in this population. These findings underscore the need for future research to understand the long-term health impacts of these interventions and for further strategies to enrich the gut microbiome of formula-fed and preterm infants.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
48
|
Snyder KB, Calkins CL, Golubkova A, Leiva T, Schlegel C, Hunter CJ. Despite Recovery from Necrotizing Enterocolitis Infants Retain a Hyperinflammatory Response to Injury. J Inflamm Res 2024; 17:331-341. [PMID: 38250141 PMCID: PMC10800089 DOI: 10.2147/jir.s436125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Background Necrotizing enterocolitis (NEC) is the leading gastrointestinal cause of death of premature neonates. NEC is associated with prematurity, a hyperinflammatory response, and dysregulation of intestinal barrier function. We hypothesize that patients with NEC will have, and continue to have after recovery, an increased hyperinflammatory intestinal response compared to those patients without NEC. Methods Neonates with NEC, those that have recovered from NEC, and those without NEC undergoing intestinal resections had specimens collected and snap frozen or generated into enteroids. The enteroids were treated with 100ug/mL lipopolysaccharide (LPS) and subjected to 24 hr of hypoxia together, then compared with untreated controls. Expression of Tumor Necrosis Factor (TNF-α) and interleukin 8 (IL-8) were evaluated via RT-qPCR and ELISA to measure inflammatory response. ANOVA determined statistical significance (p<0.05). Results There was no difference in inflammatory markers in recovered NEC tissue compared to non-NEC tissue on RTqPCR (p=0.701 TNF-α and 0.861 IL-8). However, recovered NEC enteroids demonstrate elevated levels of inflammatory markers after treatment compared to non-NEC enteroids after treatment on RTqPCR (p=0.0485 TNF-α, p=0.0057 IL-8) and ELISA (p=0.0354 TNF-α, p=0.0011 IL-8). Recovered NEC enteroids that underwent treatment demonstrated increased inflammatory markers compared to recovered NEC enteroids without treatment on RTqPCR (p=0.0045 TNF-α, p=0.0002 IL-8) and ELISA (p=0.034 TNF-α, p=0.0002 IL-8) suggesting a heightened inflammatory response to a second hit. Conclusion Intestinal tissue resected from neonates with NEC has an elevated hyperinflammatory response compared to neonates recovered from NEC and neonates without NEC. Enteroids generated from patients that have recovered from NEC have a heightened inflammatory response in response to NEC inducing stimuli compared to controls. This tendency towards an increased hyperinflammatory state may be correlated with an infant's proclivity to develop NEC and demonstrates the significance of a second hit on this tissue creating a heightened inflammatory response. This could be correlated with the impact and trajectory of an illness post recovery from NEC.
Collapse
Affiliation(s)
- Katherine B Snyder
- Division of Pediatric Surgery, Oklahoma City, OK, 73104, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Chase L Calkins
- College of Medicine, The University of Oklahoma College of Medicine, Oklahoma City, OK, 73104, USA
| | - Alena Golubkova
- Division of Pediatric Surgery, Oklahoma City, OK, 73104, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Tyler Leiva
- Division of Pediatric Surgery, Oklahoma City, OK, 73104, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | | | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma City, OK, 73104, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
49
|
Zyoud SH. Mapping the landscape of research on insulin resistance: a visualization analysis of randomized clinical trials. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:6. [PMID: 38195616 PMCID: PMC10777523 DOI: 10.1186/s41043-024-00497-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/05/2024] [Indexed: 01/11/2024]
Abstract
BACKGROUND Insulin resistance, a condition in which cells do not respond adequately to insulin, plays a crucial role in diabetes and related metabolic disorders. Randomized clinical trials (RCTs) explore interventions to manage insulin resistance, contributing to evidence-based medical progress. The current study aimed to analyze the global research landscape and trends in RCTs targeting insulin resistance. METHODS This study used bibliometric analysis and data visualization to examine RCT publications on insulin resistance from 2003 to 2022. The Scopus database was used due to its comprehensive coverage. The search strategy involved combining terms related to insulin resistance with RCT-related terms. The search query was validated, and core bibliometric indicators were used to analyze publication growth, origin, productivity, quality, and citations. RESULTS Between 2003 and 2022, 1077 RCT-focused publications on insulin resistance were identified from a pool of 24,932 related articles. The growth followed two phases, with a significant increase after 2008. The USA (n = 308; 28.60%), Iran (n = 165; 15.32%), China (n = 110; 10.21%), and the UK (n = 92; 8.54%) were the main contributors. The active institutions included Tehran University of Medical Sciences (n = 38; 3.53%) and Harvard Medical School (n = 31; 2.88%). Prominent funding agencies include the National Institutes of Health (n = 88; 8.17%) and the National Institute of Diabetes and Digestive and Kidney Diseases (n = 86; 7.99%). The top journals included the American Journal of Clinical Nutrition (n = 44; 4.09%) and Diabetes Care (n = 35; 3.25%). Co-occurrence analysis revealed three clusters addressing "utilizing lipid panels as indicators of insulin resistance," "analyzing the impact of diet composition and physical activity on insulin sensitivity among obese individuals," and "exploring insulin resistance in cases of polycystic ovary syndrome." CONCLUSIONS This comprehensive bibliometric analysis highlights the global research landscape and trends in RCTs targeting insulin resistance. Research on lipid panels, diet impact, and insulin resistance in patients with polycystic ovary syndrome will continue to be a hotspot. The findings offer valuable information on research priorities, international collaborations, and impactful publications. This study provides a foundation for future directorial investigations in this critical area of metabolic health.
Collapse
Affiliation(s)
- Sa'ed H Zyoud
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Clinical Research Centre, An-Najah National University Hospital, Nablus, 44839, Palestine.
| |
Collapse
|
50
|
Lin C, Lin Y, Wang S, Wang J, Mao X, Zhou Y, Zhang H, Chen W, Wang G. Bifidobacterium animalis subsp. lactis boosts neonatal immunity: unravelling systemic defences against Salmonella. Food Funct 2024; 15:236-254. [PMID: 38054827 DOI: 10.1039/d3fo03686c] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Bifidobacterium animalis subsp. lactis may be a useful probiotic intervention for regulating neonatal intestinal immune responses and counteracting Salmonella infection. However, recent research has focused on intestinal immunity, leaving uncertainties regarding the central, peripheral, and neural immune responses in neonates. Therefore, this study investigated the role and mechanisms of B. animalis subsp. lactis in the systemic immune responses of neonatal rats following Salmonella infection. Through extremely early pretreatment with B. animalis subsp. lactis (6 hours postnatal), the neonatal rat gut microbiota was effectively reshaped, especially the Bifidobacterium community. In the rats pretreated with B. animalis subsp. lactis, Salmonella was less prevalent in the blood, liver, spleen, and intestines following infection. The intervention promoted T lymphocyte subset balance in the spleen and thymus and fostered neurodevelopment and neuroimmune balance in the brain. Furthermore, metabolic profiling showed a strong correlation between the metabolites in the serum and colon, supporting the view that B. animalis subsp. lactis pretreatment influences the systemic immune response by modifying the composition and metabolism of the gut microbiota. Overall, the results imply that B. animalis subsp. lactis pretreatment, through the coordinated regulation of colonic and serum metabolites, influences the systemic immune responses of neonatal rats against Salmonella infection.
Collapse
Affiliation(s)
- Chunxiu Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Yugui Lin
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, Southern Medical University, Zhongshan 528400, P. R. China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Jialiang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Xuhua Mao
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi 214200, P. R. China
| | - Yonghua Zhou
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| |
Collapse
|