1
|
Graspeuntner S, Lupatsii M, van Zandbergen V, Dammann MT, Pagel J, Nguyen DN, Humberg A, Göpel W, Herting E, Rupp J, Härtel C, Fortmann I. Infants < 90 days of age with late-onset sepsis display disturbances of the microbiome-immunity interplay. Infection 2024:10.1007/s15010-024-02396-6. [PMID: 39541036 DOI: 10.1007/s15010-024-02396-6] [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: 01/23/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE We hypothesized that previously healthy infants < 90 days of age with late-onset sepsis (LOS) have disturbances of the gut microbiome with yet undefined specific immunological patterns. METHODS We performed a prospective single-center convenience sample study between January 2019 and July 2021 in a case-control design. Routine diagnostics included conventional cultures (blood, cerebrospinal fluid, urine), PCRs and inflammatory markers in infants aged < 90 days with clinical LOS. We additionally analyzed blood lymphocyte subsets including CD4 + CD25 + forkhead box protein (FoxP3)+ Tregs and performed 16 S rRNA sequencing of stool samples, both compared to age-matched healthy controls. Results were adjusted for potential confounders that may influence microbial composition. RESULTS 51 infants with fever and clinical LOS were enrolled. Bacterial sepsis was diagnosed in n = 24 (47.1%) and viral infection in n = 13 (25.5%) infants, whereas in 14 (27.3%) infants the cause of fever remained undetermined. When compared to healthy controls, the gut microbiome of LOS infants at disease onset was characterized by a shift in community composition, specifically, decreased abundance of B. longum and an increase of Bacteroidia spp. Intriguingly, the abundance of B. longum negatively correlated with the frequency of blood CD4-positive cells in healthy controls but not in infants with LOS. At one year of age, we observed microbiome differences in infants with history of LOS when compared to healthy controls, such as an increased gut microbial diversity. CONCLUSION Our data suggest potential signatures of the microbiome-immunity interplay in infants with LOS, which should be investigated further as possible targets for prevention.
Collapse
Affiliation(s)
- Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Vera van Zandbergen
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Marie-Theres Dammann
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Julia Pagel
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Ingmar Fortmann
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany.
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
2
|
Dai DLY, Petersen C, Turvey SE. Reduce, reinforce, and replenish: safeguarding the early-life microbiota to reduce intergenerational health disparities. Front Public Health 2024; 12:1455503. [PMID: 39507672 PMCID: PMC11537995 DOI: 10.3389/fpubh.2024.1455503] [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: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Socioeconomic (SE) disparity and health inequity are closely intertwined and associated with cross-generational increases in the rates of multiple chronic non-communicable diseases (NCDs) in North America and beyond. Coinciding with this social trend is an observed loss of biodiversity within the community of colonizing microbes that live in and on our bodies. Researchers have rightfully pointed to the microbiota as a key modifiable factor with the potential to ease existing health inequities. Although a number of studies have connected the adult microbiome to socioeconomic determinants and health outcomes, few studies have investigated the role of the infant microbiome in perpetuating these outcomes across generations. It is an essential and important question as the infant microbiota is highly sensitive to external forces, and observed shifts during this critical window often portend long-term outcomes of health and disease. While this is often studied in the context of direct modulators, such as delivery mode, family size, antibiotic exposure, and breastfeeding, many of these factors are tied to underlying socioeconomic and/or cross-generational factors. Exploring cross-generational socioeconomic and health inequities through the lens of the infant microbiome may provide valuable avenues to break these intergenerational cycles. In this review, we will focus on the impact of social inequality in infant microbiome development and discuss the benefits of prioritizing and restoring early-life microbiota maturation for reducing intergenerational health disparities.
Collapse
Affiliation(s)
| | | | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Kasperek MC, Velasquez Galeas A, Caetano-Silva ME, Xie Z, Ulanov A, La Frano M, Devkota S, Miller MJ, Allen JM. Microbial aromatic amino acid metabolism is modifiable in fermented food matrices to promote bioactivity. Food Chem 2024; 454:139798. [PMID: 38823201 DOI: 10.1016/j.foodchem.2024.139798] [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: 02/09/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
Ingestion of fermented foods impacts human immune function, yet the bioactive food components underlying these effects are not understood. Here, we interrogated whether fermented food bioactivity relates to microbial metabolites derived from aromatic amino acids, termed aryl-lactates. Using targeted metabolomics, we established the presence of aryl-lactates in commercially available fermented foods. After pinpointing fermented food-associated lactic acid bacteria that produce high levels of aryl-lactates, we identified fermentation conditions to increase aryl-lactate production in food matrices up to 5 × 103 fold vs. standard fermentation conditions. Using ex vivo reporter assays, we found that food matrix conditions optimized for aryl-lactate production exhibited enhanced agonist activity for the human aryl-hydrocarbon receptor (AhR) as compared to standard fermentation conditions and commercial products. Reduced microbial-induced AhR activity has emerged as a hallmark of many chronic inflammatory diseases, thus we envision strategies to enhance AhR bioactivity of fermented foods to be leveraged to improve human health.
Collapse
Affiliation(s)
- Mikaela C Kasperek
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Adriana Velasquez Galeas
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Maria Elisa Caetano-Silva
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Zifan Xie
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Alexander Ulanov
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Michael La Frano
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Suzanne Devkota
- Human Microbiome Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.
| | - Michael J Miller
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Jacob M Allen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Liu S, Pang H, Wang C, Wang Z, Wang M, Zhang Y, Zhang W, Sui Z. Rapid and accurate quantification of viable Bifidobacterium cells in milk powder with a propidium monoazide-antibiotic fluorescence in situ hybridization-flow cytometry method. J Dairy Sci 2024; 107:7678-7690. [PMID: 38908696 DOI: 10.3168/jds.2024-24876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/27/2024] [Indexed: 06/24/2024]
Abstract
Due to its beneficial effects on human health, Bifidobacterium is commonly added to milk powder. Accurate quantification of viable Bifidobacterium is essential for assessing the therapeutic efficacy of milk powder. In this study, we introduced a novel propidium monoazide (PMA)-antibiotic fluorescence in situ hybridization (AFISH)-flow cytometry (FC) method to rapidly and accurately quantify viable Bifidobacterium cells in milk powder. Briefly, Bifidobacterium cells were treated with chloramphenicol (CM) to increase their rRNA content, followed by staining with RNA-binding oligonucleotide probes, based on the AFISH technique. Then, the DNA-binding dye PMA was used to differentiate between viable and nonviable cells. The PMA-AFISH-FC method, including sample pretreatment, CM treatment, dual staining, and FC analysis, required approximately 2 h and was found to be better than the current methods. This is the first study to implement FC combined with PMA and an oligonucleotide probe for detecting Bifidobacterium.
Collapse
Affiliation(s)
- Siyuan Liu
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China; Hebei Key Laboratory of Analysis and Control for Zoonotic Pathogenic Microorganism, Hebei Agricultural University, Baoding 071001, China
| | - Huimin Pang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China; Hebei Key Laboratory of Analysis and Control for Zoonotic Pathogenic Microorganism, Hebei Agricultural University, Baoding 071001, China
| | - Chenglong Wang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China
| | - Ziquan Wang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China
| | - Meng Wang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China
| | - Yunzhe Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China; Hebei Key Laboratory of Analysis and Control for Zoonotic Pathogenic Microorganism, Hebei Agricultural University, Baoding 071001, China
| | - Wei Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China; Hebei Key Laboratory of Analysis and Control for Zoonotic Pathogenic Microorganism, Hebei Agricultural University, Baoding 071001, China.
| | - Zhiwei Sui
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China.
| |
Collapse
|
5
|
Wu J, Zhang B, Liu X, Gu W, Xu F, Wang J, Liu Q, Wang R, Hu Y, Liu J, Ji X, Lv H, Li X, Peng L, Li X, Zhang Y, Wang S. An Intelligent Intestine-on-a-Chip for Rapid Screening of Probiotics with Relief-Enteritis Function. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408485. [PMID: 39344562 DOI: 10.1002/adma.202408485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Screening probiotics with specific functions is essential for advancing probiotic research. Current screening methods primarily use animal studies or clinical trials, which are inefficient and costly in terms of time, money, and labor. An intelligent intestine-on-a-chip integrating machine learning (ML) is developed to screen relief-enteritis functional probiotics. A high-throughput microfluidic chip combined with environment control systems provides a standardized and scalable intestinal microenvironment for multiple probiotic cocultures. An unsupervised ML-based score analyzer is constructed to accurately, comprehensively, and efficiently evaluate interactions between 12 Bifidobacterium strains and host cells of the colitis model in the intestine-on-a-chips. The most effective contender, Bifidobacterium longum 3-14, is discovered to relieve intestinal inflammation and enhance epithelial barrier function in vitro and in vivo. A distinct advantage of this strategy is that it can intelligently differentiate small therapeutic variations in probiotic strains and prioritize their efficacies, allowing for economical, efficient, accurate functional probiotics screening.
Collapse
Affiliation(s)
- Jing Wu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaoxia Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Wentao Gu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Fupei Xu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qisijing Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ruican Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yaozhong Hu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jingmin Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xuemeng Ji
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huan Lv
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xinyang Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lijun Peng
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiang Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yan Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shuo Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
6
|
Belchik SE, Oba PM, Lin CY, Swanson KS. Effects of a Veterinary Gastrointestinal Low-Fat Diet on Fecal Characteristics, Metabolites, and Microbiota Concentrations of Adult Dogs Treated with Metronidazole. J Anim Sci 2024; 102:skae297. [PMID: 39344678 PMCID: PMC11568346 DOI: 10.1093/jas/skae297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024] Open
Abstract
Antibiotics are known to cause loose stools, disrupt the fecal microbiota, and alter fecal bile acid (BA) profiles of dogs. Recovery may be aided by diet, but little research has been conducted. The objective of this study was to determine how a veterinary low-fat diet affected the fecal characteristics, metabolites, BA, and microbiota of dogs receiving antibiotics. Twenty-four healthy adult dogs [7.38 ± 1.95 yr; 7.67 ± 0.76 kg body weight (BW)] were used in an 8-wk completely randomized design study. During a 2-wk baseline, all dogs were fed a leading grocery brand dry kibble diet (GBD). Over the next 2 wk, dogs were fed GBD and received metronidazole orally (20 mg/kg BW twice daily). At wk 4, dogs were randomly allotted to one of two treatments [GBD or Blue Buffalo Natural Veterinary Diet GI Gastrointestinal Support Low-Fat (BB)] and fed for 4 wk. Fecal scores were recorded daily and fresh fecal samples were collected at wk 2, 4, 5, 6, 7, and 8 for measurement of pH, dry matter content, and metabolite and BA concentrations. Fecal microbiota populations were analyzed by 16S rRNA gene amplicon sequencing and qPCR-based dysbiosis index (DI). All data were analyzed as repeated measures using the Mixed Models procedure of SAS 9.4, testing for effects of treatment, time, and treatment*time and significance set at P<0.05. Metronidazole increased (P<0.0001) fecal scores (looser stools), reduced fecal short-chain fatty acid, branched-chain fatty acid, phenol, and indole concentrations, increased primary BA concentrations, and decreased secondary BA concentrations. Metronidazole also reduced fecal bacterial alpha diversity, altered the abundance of 58 bacterial genera, and increased DI. During antibiotic recovery, change in fecal pH, dry matter percentage, and metabolite and immunoglobulin A concentrations were altered (P<0.05) by diet. Fecal BA concentrations recovered quickly for all dogs. Change in lithocholic acid was affected (P<0.0001) by diet, but other BA were not. Recovery of over 25 bacterial genera was impacted by diet (P<0.05). While many bacterial taxa returned to baseline levels after 4 wk, others did not fully recover. DI and bacterial alpha diversity measures recovered quickly for all dogs, but were not impacted by diet. In conclusion, metronidazole drastically altered the fecal microbiota and metabolites of dogs. While most variables returned to baseline by wk 8, diet may be used to aid in recovery.
Collapse
Affiliation(s)
- Sara E Belchik
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Patricia M Oba
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
7
|
Zhao J, Guo Y, Jiang Q, Lan H, Hung WL, Lynch B. Bifidobacterium longum subsp. infantis YLGB-1496-Toxicological evaluation. J Appl Toxicol 2024. [PMID: 39252460 DOI: 10.1002/jat.4688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/11/2024]
Abstract
Bifidobacterium infantis YLGB-1496, originally isolated from breast milk from a Taiwanese mother, is under study for use as a probiotic. As part of safety assessment, an Ames, in vivo mouse micronucleus, and in vivo mouse spermatocyte chromosome aberration assay were conducted along with a 13-week oral rat toxicity study. B. infantis YLGB-1496 had no activity in any of the genotoxicity assays. Administration of the bacteria to Sprague-Dawley rats at doses ranging from 0 to 1.5 g/kg bw/day had no treatment-related effects on any of the endpoints measured. There appear to be no concerns for translocation or pathogenicity of B. infantis YLGB-1496 based on extensive experience with the species in general. The results of the current investigations support potential use of B. infantis YLGB-1496 as a probiotic in infant formula.
Collapse
Affiliation(s)
- Jian Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yueyi Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qiuyue Jiang
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Barry Lynch
- Intertek Health Sciences Inc., Mississauga, ON, Canada
| |
Collapse
|
8
|
Retuerto M, Al-Shakhshir H, Herrada J, McCormick TS, Ghannoum MA. Analysis of Gut Bacterial and Fungal Microbiota in Children with Autism Spectrum Disorder and Their Non-Autistic Siblings. Nutrients 2024; 16:3004. [PMID: 39275319 PMCID: PMC11396985 DOI: 10.3390/nu16173004] [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: 08/02/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a multifactorial disorder involving genetic and environmental factors leading to pathophysiologic symptoms and comorbidities including neurodevelopmental disorders, anxiety, immune dysregulation, and gastrointestinal (GI) abnormalities. Abnormal intestinal permeability has been reported among ASD patients and it is well established that disturbances in eating patterns may cause gut microbiome imbalance (i.e., dysbiosis). Therefore, studies focusing on the potential relationship between gut microbiota and ASD are emerging. We compared the intestinal bacteriome and mycobiome of a cohort of ASD subjects with their non-ASD siblings. Differences between ASD and non-ASD subjects include a significant decrease at the phylum level in Cyanobacteria (0.015% vs. 0.074%, p < 0.0003), and a significant decrease at the genus level in Bacteroides (28.3% vs. 36.8%, p < 0.03). Species-level analysis showed a significant decrease in Faecalibacterium prausnitzii, Prevotella copri, Bacteroides fragilis, and Akkermansia municiphila. Mycobiome analysis showed an increase in the fungal Ascomycota phylum (98.3% vs. 94%, p < 0.047) and an increase in Candida albicans (27.1% vs. 13.2%, p < 0.055). Multivariate analysis showed that organisms from the genus Delftia were predictive of an increased odds ratio of ASD, whereas decreases at the phylum level in Cyanobacteria and at the genus level in Azospirillum were associated with an increased odds ratio of ASD. We screened 24 probiotic organisms to identify strains that could alter the growth patterns of organisms identified as elevated within ASD subject samples. In a preliminary in vivo preclinical test, we challenged wild-type Balb/c mice with Delftia acidovorans (increased in ASD subjects) by oral gavage and compared changes in behavioral patterns to sham-treated controls. An in vitro biofilm assay was used to determine the ability of potentially beneficial microorganisms to alter the biofilm-forming patterns of Delftia acidovorans, as well as their ability to break down fiber. Downregulation of cyanobacteria (generally beneficial for inflammation and wound healing) combined with an increase in biofilm-forming species such as D. acidovorans suggests that ASD-related GI symptoms may result from decreases in beneficial organisms with a concomitant increase in potential pathogens, and that beneficial probiotics can be identified that counteract these changes.
Collapse
Affiliation(s)
- Mauricio Retuerto
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hilmi Al-Shakhshir
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Janet Herrada
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mahmoud A Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for Medical Mycology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
9
|
Shen X, Ma C, Yang Y, Liu X, Wang B, Wang Y, Zhang G, Bian X, Zhang N. The Role and Mechanism of Probiotics Supplementation in Blood Glucose Regulation: A Review. Foods 2024; 13:2719. [PMID: 39272484 PMCID: PMC11394447 DOI: 10.3390/foods13172719] [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: 08/02/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
With economic growth and improved living standards, the incidence of metabolic diseases such as diabetes mellitus caused by over-nutrition has risen sharply worldwide. Elevated blood glucose and complications in patients seriously affect the quality of life and increase the economic burden. There are limitations and side effects of current hypoglycemic drugs, while probiotics, which are safe, economical, and effective, have good application prospects in disease prevention and remodeling of intestinal microecological health and are gradually becoming a research hotspot for diabetes prevention and treatment, capable of lowering blood glucose and alleviating complications, among other things. Probiotic supplementation is a microbiologically based approach to the treatment of type 2 diabetes mellitus (T2DM), which can achieve anti-diabetic efficacy through the regulation of different tissues and metabolic pathways. In this study, we summarize recent findings that probiotic intake can achieve blood glucose regulation by modulating intestinal flora, decreasing chronic low-grade inflammation, modulating glucagon-like peptide-1 (GLP-1), decreasing oxidative stress, ameliorating insulin resistance, and increasing short-chain fatty acids (SCFAs) content. Moreover, the mechanism, application, development prospect, and challenges of probiotics regulating blood glucose were discussed to provide theoretical references and a guiding basis for the development of probiotic preparations and related functional foods regulating blood glucose.
Collapse
Affiliation(s)
- Xinyu Shen
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Chunmin Ma
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Yang Yang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xiaofei Liu
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Bing Wang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Yan Wang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Guang Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xin Bian
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Na Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| |
Collapse
|
10
|
Liu L, He G, Yu R, Lin B, Lin L, Wei R, Zhu Z, Xu Y. Causal relationships between gut microbiome and obstructive sleep apnea: a bi-directional Mendelian randomization. Front Microbiol 2024; 15:1410624. [PMID: 39309525 PMCID: PMC11414551 DOI: 10.3389/fmicb.2024.1410624] [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: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 09/25/2024] Open
Abstract
Background Previous studies have identified a clinical association between gut microbiota and Obstructive sleep apnea (OSA), but the potential causal relationship between the two has not been determined. Therefore, we aim to utilize Mendelian randomization (MR) to investigate the potential causal effects of gut microbiota on OSA and the impact of OSA on altering the composition of gut microbiota. Methods Bi-directional MR and replicated validation were utilized. Summary-level genetic data of gut microbiota were derived from the MiBioGen consortium and the Dutch Microbiome Project (DMP). Summary statistics of OSA were drawn from FinnGen Consortium and Million Veteran Program (MVP). Inverse-variance-weighted (IVW), weighted median, MR-Egger, Simple Mode, and Weighted Mode methods were used to evaluate the potential causal link between gut microbiota and OSA. Results We identified potential causal associations between 23 gut microbiota and OSA. Among them, genus Eubacterium xylanophilum group (OR = 0.86; p = 0.00013), Bifidobacterium longum (OR = 0.90; p = 0.0090), Parabacteroides merdae (OR = 0.85; p = 0.00016) retained a strong negative association with OSA after the Bonferroni correction. Reverse MR analyses indicated that OSA was associated with 20 gut microbiota, among them, a strong inverse association between OSA and genus Anaerostipes (beta = -0.35; p = 0.00032) was identified after Bonferroni correction. Conclusion Our study implicates the potential bi-directional causal effects of the gut microbiota on OSA, potentially providing new insights into the prevention and treatment of OSA through specific gut microbiota.
Collapse
Affiliation(s)
- Liangfeng Liu
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Guanwen He
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Rong Yu
- Department of Pediatrics, Jiaocheng District Maternal and Child Health Hospital, Ningde, Fujian, China
| | - Bingbang Lin
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Liangqing Lin
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Rifu Wei
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Zhongshou Zhu
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
| | - Yangbin Xu
- Department of Otolaryngology, Head and Neck Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian, China
- Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Zhang H, Wang J, Shen J, Chen S, Yuan H, Zhang X, Liu X, Yu Y, Li X, Gao Z, Wang Y, Wang J, Song M. Prophylactic supplementation with Bifidobacterium infantis or its metabolite inosine attenuates cardiac ischemia/reperfusion injury. IMETA 2024; 3:e220. [PMID: 39135700 PMCID: PMC11316933 DOI: 10.1002/imt2.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 08/15/2024]
Abstract
Emerging evidence has demonstrated the profound impact of the gut microbiome on cardiovascular diseases through the production of diverse metabolites. Using an animal model of myocardial ischemia-reperfusion (I/R) injury, we found that the prophylactic administration of a well-known probiotic, Bifidobacterium infantis (B. infantis), exhibited cardioprotective effects in terms of preserving cardiac contractile function and preventing adverse cardiac remodeling following I/R and that these cardioprotective effects were recapitulated by its metabolite inosine. Transcriptomic analysis further revealed that inosine mitigated I/R-induced cardiac inflammation and cell death. Mechanistic investigations elucidated that inosine suppressed the production of pro-inflammatory cytokines and reduced the numbers of dendritic cells and natural killer cells, achieved through the activation of the adenosine A2A receptor (A2AR) that when inhibited abrogated the cardioprotective effects of inosine. Additionally, in vitro studies using C2C12 myoblasts revealed that inosine attenuated cell death by serving as an alternative carbon source for adenosine triphosphate (ATP) generation through the purine salvage pathway when subjected to oxygen-glucose deprivation/reoxygenation that simulated myocardial I/R injury. Likewise, inosine reversed the I/R-induced decrease in ATP levels in mouse hearts. Taken together, our findings indicate that B. infantis or its metabolite inosine exerts cardioprotective effects against I/R by suppressing cardiac inflammation and attenuating cardiac cell death, suggesting prophylactic therapeutic options for acute ischemic cardiac injury.
Collapse
Affiliation(s)
- Hao Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiawan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Department of AnesthesiologyBeijing Chao‐Yang HospitalBeijingChina
| | - Jianghua Shen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Siqi Chen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Hailong Yuan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifengChina
| | - Xuan Zhang
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Xu Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Ying Yu
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Xinran Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Zeyu Gao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifengChina
| | - Jun Wang
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Moshi Song
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
12
|
Lai C, Huang L, Wang Y, Huang C, Luo Y, Qin X, Zeng J. Effect of different delivery modes on intestinal microbiota and immune function of neonates. Sci Rep 2024; 14:17452. [PMID: 39075163 PMCID: PMC11286838 DOI: 10.1038/s41598-024-68599-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024] Open
Abstract
Different delivery methods can cause variations in the composition and structure of intestinal microbiota in neonates. However, the impact of the microecological environment on host immune function requires further investigation. In this study, 75 healthy neonates were divided into two groups: vaginal delivery group (n = 36) and cesarean section group (n = 39). Fecal and peripheral blood samples were collected from the 7th to the 10th day. 16S rRNA sequencing technique was performed to investigate the gut microbiota on fecal samples. Levels of immunoglobulins and Th1 and Th2 cells in the peripheral blood of neonates were measured. The abundance of Escherichia, Bifidobacterium, and Bacteroides in neonates in the cesarean section group was significantly lower than that in the vaginal delivery group. Metabolic pathway analysis showed three significantly up-regulated metabolic pathways in the intestinal microbiota of neonates in the cesarean section group. The levels of serum IgG and IL-12p70 in the cesarean section group were lower than those in the vaginal delivery group, and the proportion of IFN-γ/IL-4 was significantly lower in the cesarean section group compared to the vaginal delivery group. The mode of delivery has potential impact on the intestinal microbiota and immune functions of neonates, potentially leading to an imbalance of Th1/Th2 cells in neonates delivered by cesarean section.
Collapse
Affiliation(s)
- Chunhui Lai
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China
| | - Li Huang
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China
| | - Yijin Wang
- Department of Paediatrics, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chaosheng Huang
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China
| | - Yibing Luo
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China
| | - Xuemei Qin
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China
| | - Jianghui Zeng
- Department of Clinical Laboratory, The Second Nanning People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Molecular Immunology Research, Nanning, Guangxi, China.
| |
Collapse
|
13
|
Liu Y, Zhang Y, Guo C, Li M, Wang Y, Zhang L. Analysis of gut microecological characteristics and differences between children with biliary atresia and non-biliary atresia in infantile cholestasis. Front Cell Infect Microbiol 2024; 14:1402329. [PMID: 38947125 PMCID: PMC11212454 DOI: 10.3389/fcimb.2024.1402329] [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: 03/22/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction In infants with cholestasis, variations in the enterohepatic circulation of bile acids and the gut microbiota (GM) characteristics differ between those with biliary atresia (BA) and non-BA, prompting a differential analysis of their respective GM profiles. Methods Using 16S rDNA gene sequencing to analyse the variance in GM composition among three groups: infants with BA (BA group, n=26), non-BA cholestasis (IC group, n=37), and healthy infants (control group, n=50). Additionally, correlation analysis was conducted between GM and liver function-related indicators. Results Principal component analysis using Bray-Curtis distance measurement revealed a significant distinction between microbial samples in the IC group compared to the two other groups. IC-accumulated co-abundance groups exhibited positive correlations with aspartate aminotransferase, alanine aminotransferase, total bilirubin, direct bilirubin, and total bile acid serum levels. These correlations were notably reinforced upon the exclusion of microbial samples from children with BA. Conclusion The varying "enterohepatic circulation" status of bile acids in children with BA and non-BA cholestasis contributes to distinct GM structures and functions. This divergence underscores the potential for targeted GM interventions tailored to the specific aetiologies of cholestasis.
Collapse
Affiliation(s)
- Yajun Liu
- Department of Pediatrics, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Yuan Zhang
- Department of Pediatrics, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Cheng Guo
- Department of Pediatrics, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Muxia Li
- Office of Academic Research, Beijing Children’s Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ye Wang
- Department of Pediatrics, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Lin Zhang
- Department of Pediatrics, Hebei Medical University Third Hospital, Shijiazhuang, China
| |
Collapse
|
14
|
Bellomo AR, Rotondi G, Rago P, Bloise S, Di Ruzza L, Zingoni A, Di Valerio S, Valzano E, Di Pierro F, Cazzaniga M, Bertuccioli A, Guasti L, Zerbinati N, Lubrano R. Effect of Bifidobacterium bifidum Supplementation in Newborns Born from Cesarean Section on Atopy, Respiratory Tract Infections, and Dyspeptic Syndromes: A Multicenter, Randomized, and Controlled Clinical Trial. Microorganisms 2024; 12:1093. [PMID: 38930475 PMCID: PMC11205812 DOI: 10.3390/microorganisms12061093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Cesarean section is considered a possible trigger of atopy and gut dysbiosis in newborns. Bifidobacteria, and specifically B. bifidum, are thought to play a central role in reducing the risk of atopy and in favoring gut eubiosis in children. Nonetheless, no trial has ever prospectively investigated the role played by this single bacterial species in preventing atopic manifestations in children born by cesarean section, and all the results published so far refer to mixtures of probiotics. We have therefore evaluated the impact of 6 months of supplementation with B. bifidum PRL2010 on the incidence, in the first year of life, of atopy, respiratory tract infections, and dyspeptic syndromes in 164 children born by cesarean (versus 249 untreated controls). The results of our multicenter, randomized, and controlled trial have shown that the probiotic supplementation significantly reduced the incidence of atopic dermatitis, upper and lower respiratory tract infections, and signs and symptoms of dyspeptic syndromes. Concerning the gut microbiota, B. bifidum supplementation significantly increased α-biodiversity and the relative values of the phyla Bacteroidota and Actinomycetota, of the genus Bacteroides, Bifidobacterium and of the species B. bifidum and reduced the relative content of Escherichia/Shigella and Haemophilus. A 6-month supplementation with B. bifidum in children born by cesarean section reduces the risk of gut dysbiosis and has a positive clinical impact that remains observable in the following 6 months of follow-up.
Collapse
Affiliation(s)
- Anna Rita Bellomo
- Dipartimento Materno Infantile e di Scienze Urologiche, Sapienza Università di Roma, UOC di Pediatria e Neonatologia-Polo Pontino, 04100 Latina, Italy; (A.R.B.); (P.R.)
| | - Giulia Rotondi
- Pediatric Surgery Unit, Gaslini Children Hospital and Research Institute, 16147 Genoa, Italy
| | - Prudenza Rago
- Dipartimento Materno Infantile e di Scienze Urologiche, Sapienza Università di Roma, UOC di Pediatria e Neonatologia-Polo Pontino, 04100 Latina, Italy; (A.R.B.); (P.R.)
| | - Silvia Bloise
- Dipartimento Materno Infantile e di Scienze Urologiche, Sapienza Università di Roma, UOC di Pediatria e Neonatologia-Polo Pontino, 04100 Latina, Italy; (A.R.B.); (P.R.)
| | - Luigi Di Ruzza
- UOC Pediatria e Nido, Ospedale S.S. Trinità, 03039 Sora, Italy
| | - Annamaria Zingoni
- UOC Pediatria e Neonatologia, Ospedale G.B. Grassi, 00122 Ostia, Italy
| | - Susanna Di Valerio
- UOC Neonatologia e Terapia Intensiva Neonatale, Ospedale S. Spirito, 65124 Pescara, Italy
| | - Eliana Valzano
- UOC Neonatologia e Terapia Intensiva Neonatale, Ospedale S. Spirito, 65124 Pescara, Italy
| | - Francesco Di Pierro
- Scientific & Research Department, Velleja Research, 20125 Milan, Italy
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | | | - Alexander Bertuccioli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Riccardo Lubrano
- Dipartimento Materno Infantile e di Scienze Urologiche, Sapienza Università di Roma, UOC di Pediatria e Neonatologia-Polo Pontino, 04100 Latina, Italy; (A.R.B.); (P.R.)
| |
Collapse
|
15
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
16
|
Tang J, Zhao M, Miao X, Chen H, Zhao B, Wang Y, Guo Y, Wang T, Cheng X, Ruan H, Zhang J. Bifidobacterium longum GL001 alleviates rat intestinal ischemia-reperfusion injury by modulating gut microbiota composition and intestinal tissue metabolism. Food Funct 2024; 15:3653-3668. [PMID: 38487897 DOI: 10.1039/d3fo03669c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Intestinal ischemia-reperfusion (IIR) injury leads to inflammation and oxidative stress, resulting in intestinal barrier damage. Probiotics, due to their anti-inflammatory and antioxidant properties, are considered for potential intervention to protect the intestinal barrier during IIR injury. Bifidobacterium longum, a recognized probiotic, has targeted effects on IIR injury, but its mechanisms of action are not yet understood. To investigate the mechanism of Bifidobacterium longum intervention in IIR injury, we conducted a study using a rat IIR injury model. The results showed that Bifidobacterium longum could alleviate inflammation and oxidative stress induced by IIR injury by suppressing the NF-κB inflammatory pathway and activating the Keap1/Nrf2 signaling pathway. Bifidobacterium longum GL001 also increased the abundance of the gut microbiota such as Oscillospira, Ouminococcus, Corynebacterium, Lactobacillus, and Akkermansia, while decreasing the abundance of Allobaculum, [Prevotella], Bacteroidaceae, Bacteroides, Shigella, and Helicobacter. In addition, Bifidobacterium longum GL001 reversed the changes in amino acids and bile acids induced by IIR injury and reduced the levels of DL-cysteine, an oxidative stress marker, in intestinal tissue. Spearman correlation analysis showed that L-cystine was positively correlated with Lactobacillus and negatively correlated with Shigella, while DL-proline was positively correlated with Akkermansia. Moreover, bile acids, cholic acid and lithocholic acid, were negatively correlated with Lactobacillus and positively correlated with Shigella. Therefore, Bifidobacterium longum GL001 may alleviate IIR injury by regulating the gut microbiota to modulate intestinal lipid peroxidation and bile acid metabolism.
Collapse
Affiliation(s)
- Jilang Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Mingchao Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Xue Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Hong Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Binger Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Yingying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Yingchao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Tiantian Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Xin Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Hongri Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| |
Collapse
|
17
|
Smulders T, Van Der Schee MP, Maitland-Van Der Zee AH, Dikkers FG, Van Drunen CM. Influence of the gut and airway microbiome on asthma development and disease. Pediatr Allergy Immunol 2024; 35:e14095. [PMID: 38451070 DOI: 10.1111/pai.14095] [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: 12/14/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024]
Abstract
There are ample data to suggest that early-life dysbiosis of both the gut and/or airway microbiome can predispose a child to develop along a trajectory toward asthma. Although individual studies show clear associations between dysbiosis and asthma development, it is less clear what (collection of) bacterial species is mechanistically responsible for the observed effects. This is partly due to issues related to the asthma diagnosis and the broad spectrum of anatomical sites, sample techniques, and analysis protocols that are used in different studies. Moreover, there is limited attention for potential differences in the genetics of individuals that would affect the outcome of the interaction between the environment and that individual. Despite these challenges, the first bacterial components were identified that are able to affect the transcriptional state of human cells, ergo the immune system. Such molecules could in the future be the basis for intervention studies that are now (necessarily) restricted to a limited number of bacterial species. For this transition, it might be prudent to develop an ex vivo human model of a local mucosal immune system to better and safer explore the impact of such molecules. With this approach, we might move beyond association toward understanding of causality.
Collapse
Affiliation(s)
- Tamar Smulders
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Marc P Van Der Schee
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Anke H Maitland-Van Der Zee
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Frederik G Dikkers
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Cornelis M Van Drunen
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
Paterniti I, Scuderi SA, Cambria L, Nostro A, Esposito E, Marino A. Protective Effect of Probiotics against Pseudomonas aeruginosa Infection of Human Corneal Epithelial Cells. Int J Mol Sci 2024; 25:1770. [PMID: 38339047 PMCID: PMC10855269 DOI: 10.3390/ijms25031770] [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: 12/31/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Probiotic therapy needs consideration as an alternative strategy to prevent and possibly treat corneal infection. This study aimed to assess the preventive effect of Lactobacillus reuteri and Bifidobacterium longum subsp. infantis on reducing the infection of human corneal epithelial (HCE) cells caused by Pseudomonas aeruginosa. The probiotics' preventive effect against infection was evaluated in cell monolayers pretreated with each probiotic 1 h and 24 h prior to P. aeruginosa challenge followed by 1 h and 24 h of growth in combination. Cell adhesion, cytotoxicity, anti-inflammatory, and antinitrosative activities were evaluated. L. reuteri and B. longum adhered to HCE cells, preserved occludin tight junctions' integrity, and increased mucin production on a SkinEthicTM HCE model. Pretreatment with L. reuteri or B. longum significantly protected HCE cells from infection at 24 h, increasing cell viability at 110% (110.51 ± 5.15; p ≤ 0.05) and 137% (137.55 ± 11.97; p ≤ 0.05), respectively. Each probiotic showed anti-inflammatory and antinitrosative activities, reducing TNF-α level (p ≤ 0.001) and NOx amount (p ≤ 0.001) and reestablishing IL-10 level (p ≤ 0.001). In conclusion, this study demonstrated that L. reuteri and B. longum exert protective effects in the context of corneal infection caused by P. aeruginosa by restoring cell viability and modulating inflammatory cytokine release.
Collapse
Affiliation(s)
| | | | | | | | | | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (I.P.); (S.A.S.); (L.C.); (A.N.); (E.E.)
| |
Collapse
|
19
|
Ding M, Li B, Chen H, Ross RP, Stanton C, Jiang S, Zhao J, Chen W, Yang B. Bifidobacterium longum subsp. infantis regulates Th1/Th2 balance through the JAK-STAT pathway in growing mice. MICROBIOME RESEARCH REPORTS 2024; 3:16. [PMID: 38841405 PMCID: PMC11149089 DOI: 10.20517/mrr.2023.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 06/07/2024]
Abstract
Objectives: Bifidobacterium longum subsp. infantis is a dominant bacterium in infant gut, which plays a critical role in maintaining the health and development of infants. This study investigated the abilities of eight different strains of B. longum subsp. infantis to regulate the T helper (Th)1/Th2 balance. Methods: Eight B. longum subsp. infantis strains, including I2MI (FJSWXI2MIM1), I4MI [FJSWXI4MI (CCFM1270)], I4MNI (FJSWXI4MNIM1), I5TI (FJSWXI5TIM1), I6TI (FJSWXI6TIM1), I8TI [FJSWXI8TI (CCFM1271)], I10TI [FJSWXI10TI (CCFM1272)], and B6MNI [BJSWXB6MNIM1 (CCFM1269)], were gavaged to BALB/C pups in both female (n = 8) and male (n = 8) mice starting from 1 to 3 weeks old (1 × 109 CFU/day/mice). Selected immune cells were assessed by immunofluorescence and flow cytometry. Cytokines and immunoglobulins were determined by ELISA. Bacterial and bifidobacterial communities were determined by 16S rRNA gene sequencing and bifidobacterial groEL sequencing. Results: B. longum subsp. infantis I4MI and I8TI were shown to increase the ration of colonic IgG2a/IgE in male mice (P < 0.05). B6MNI was demonstrated to significantly increase the levels of colonic IFN-γ and IgG2a, as well as the ratio of IgG2a/IgE in female mice (P < 0.05). It was also shown to significantly increase the ratio of colonic IgG2a/IgE (P < 0.05) and reduce the level of colonic IL-4 in male mice (P < 0.05). Furthermore, B6MNI was demonstrated to regulate colonic JAK/STAT pathway in both male and female mice. I4MI, I5TI, and B6MNI were shown to increase the relative abundance of Bifidobacterium and B. longum subsp. infantis in both male and female mice, whereas I8TI was only shown to increase the relative abundance of Bifidobacterium and B. longum subsp. infantis in male mice (P < 0.05). Conclusion: These results indicated supplementation with B. longum subsp. infantis in early infancy may regulate the Th1/Th2 immune balance, which may prevent the development of related diseases.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - R. Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork T12 R229, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork T12 R229, Ireland
- Teagasc Food Research Centre, Moorepark, Co. Cork P61 C996, Ireland
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100015, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
20
|
Shen X, Xie A, Li Z, Jiang C, Wu J, Li M, Yue X. Research Progress for Probiotics Regulating Intestinal Flora to Improve Functional Dyspepsia: A Review. Foods 2024; 13:151. [PMID: 38201179 PMCID: PMC10778471 DOI: 10.3390/foods13010151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Functional dyspepsia (FD) is a common functional gastrointestinal disorder. The pathophysiology remains poorly understood; however, alterations in the small intestinal microbiome have been observed. Current treatments for FD with drugs are limited, and there are certain safety problems. A class of active probiotic bacteria can control gastrointestinal homeostasis, nutritional digestion and absorption, and the energy balance when taken in certain dosages. Probiotics play many roles in maintaining intestinal microecological balance, improving the intestinal barrier function, and regulating the immune response. The presence and composition of intestinal microorganisms play a vital role in the onset and progression of FD and serve as a critical factor for both regulation and potential intervention regarding the management of this condition. Thus, there are potential advantages to alleviating FD by regulating the intestinal flora using probiotics, targeting intestinal microorganisms. This review summarizes the research progress of probiotics regarding improving FD by regulating intestinal flora and provides a reference basis for probiotics to improve FD.
Collapse
Affiliation(s)
- Xinyu Shen
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (X.S.); (Z.L.); (C.J.); (J.W.)
| | - Aijun Xie
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 119077, Singapore;
| | - Zijing Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (X.S.); (Z.L.); (C.J.); (J.W.)
| | - Chengxi Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (X.S.); (Z.L.); (C.J.); (J.W.)
| | - Jiaqi Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (X.S.); (Z.L.); (C.J.); (J.W.)
| | - Mohan Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China; (X.S.); (Z.L.); (C.J.); (J.W.)
| | - Xiqing Yue
- Shenyang Key Laboratory of Animal Product Processing, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
21
|
Falzone L, Lavoro A, Candido S, Salmeri M, Zanghì A, Libra M. Benefits and concerns of probiotics: an overview of the potential genotoxicity of the colibactin-producing Escherichia coli Nissle 1917 strain. Gut Microbes 2024; 16:2397874. [PMID: 39229962 PMCID: PMC11376418 DOI: 10.1080/19490976.2024.2397874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Recently, the mounting integration of probiotics into human health strategies has gathered considerable attention. Although the benefits of probiotics have been widely recognized in patients with gastrointestinal disorders, immune system modulation, and chronic-degenerative diseases, there is a growing need to evaluate their potential risks. In this context, new concerns have arisen regarding the safety of probiotics as some strains may have adverse effects in humans. Among these strains, Escherichia coli Nissle 1917 (EcN) exhibited traits of concern due to a pathogenic locus in its genome that produces potentially genotoxic metabolites. As the use of probiotics for therapeutic purposes is increasing, the effects of potentially harmful probiotics must be carefully evaluated. To this end, in this narrative review article, we reported the findings of the most relevant in vitro and in vivo studies investigating the expanding applications of probiotics and their impact on human well-being addressing concerns arising from the presence of antibiotic resistance and pathogenic elements, with a focus on the polyketide synthase (pks) pathogenic island of EcN. In this context, the literature data here discussed encourages a thorough profiling of probiotics to identify potential harmful elements as done for EcN where potential genotoxic effects of colibactin, a secondary metabolite, were observed. Specifically, while some studies suggest EcN is safe for gastrointestinal health, conflicting findings highlight the need for further research to clarify its safety and optimize its use in therapy. Overall, the data here presented suggest that a comprehensive assessment of the evolving landscape of probiotics is essential to make evidence-based decisions and ensure their correct use in humans.
Collapse
Affiliation(s)
- Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| | - Mario Salmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonino Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology 'G.F. Ingrassia', University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| |
Collapse
|
22
|
Zhang G, He M, Xiao L, Jiao Y, Han J, Li C, Miller MJ, Zhang L. Milk fat globule membrane protects Bifidobacterium longum ssp. infantis ATCC 15697 against bile stress by modifying global transcriptional responses. J Dairy Sci 2024; 107:91-104. [PMID: 37678788 DOI: 10.3168/jds.2023-23591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023]
Abstract
The milk fat globule membrane (MFGM) can protect probiotic bacteria from bile stress. However, its potential mechanism has not been reported. In this study, the viability, morphology and gene transcriptional response of Bifidobacterium longum ssp. infantis ATCC 15697 (BI_15697) stressed by bile salts with or without MFGM were investigated. It was shown that MFGM alleviated the reduction in BI_15697 population induced by 0.2% porcine bile stress and restored the population to the control levels. MFGM ameliorated the shrunken, fragmented appearance and irregular morphology of BI_15697 and maintained cell integrity disrupted by bile stress. RNA-sequencing results showed that MFGM increased transport of glucose and raffinose and decreased that of branched-chain amino acids (BCAA) in the presence of bile salts. MFGM stimulated the expression of genes involved in the synthesis of raffinose in galactose metabolism and the metabolism of BCAA, suggesting that MFGM stimulated the accumulation of raffinose and BCAA in the presence of bile. In addition, MFGM stimulated the expression of 2 bile efflux transporters under bile stress. Together, the multifactorial response helps BI_15697 excrete bile salts and maintain cellular integrity in response to bile stress. This study proposes a mechanism for the protection of BI_15697 against bile salt stress by MFGM, thereby providing a molecular basis for its application in incorporation of probiotics.
Collapse
Affiliation(s)
- Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Mingxue He
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lihong Xiao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Jianchun Han
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150030, China.
| | - Chun Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Michael J Miller
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
23
|
Raslan MA, Raslan SA, Shehata EM, Mahmoud AS, Viana MVC, Barh D, Sabri NA, Azevedo V. Applications of Proteomics in Probiotics Having Anticancer and Chemopreventive Properties. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:243-256. [PMID: 38409425 DOI: 10.1007/978-3-031-50624-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Proteomics has grown in importance in molecular sciences because it gives vital information on protein identification, expression levels, and alteration. Cancer is one of the world's major causes of death and is the major focus of much research. Cancer risk is determined by hereditary variables as well as the body's immunological condition. Probiotics have increasing medical importance due to their therapeutic influence on the human body in the prevention and treatment of numerous chronic illnesses, including cancer, with no adverse effects. Several anticancer, anti-inflammatory, and chemopreventive probiotics are studied using different proteomic approaches like two-dimensional gel electrophoresis, liquid chromatography-mass spectrometry, and matrix-assisted laser desorption/ionization mass spectrometry. To gain relevant information about probiotic characteristics, data from the proteomic analysis are evaluated and processed using bioinformatics pipelines. Proteomic studies showed the significance of different proteomic approaches in characterization, comparing strains, and determination of oxidative stress of different probiotics. Moreover, proteomic approaches identified different proteins that are involved in glucose metabolism and the formation of cell walls or cell membranes, and the differences in the expression of critical enzymes in the HIF-1 signaling pathway, starch, and sucrose metabolism, and other critical metabolic pathways.
Collapse
Affiliation(s)
| | | | | | - Amr S Mahmoud
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marcus Vinicius Canário Viana
- Laboratório de Genética Celular e Molecular, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Debmalya Barh
- Laboratório de Genética Celular e Molecular, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
24
|
Zhou Y, Sheng YJ, Li CY, Zou L, Tong CY, Zhang Y, Cao G, Shou D. Beneficial effect and mechanism of natural resourced polysaccharides on regulating bone metabolism through intestinal flora: A review. Int J Biol Macromol 2023; 253:127428. [PMID: 37838110 DOI: 10.1016/j.ijbiomac.2023.127428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
Bone metabolism is an important biological process for maintaining bone health. Polysaccharides of natural origin exert beneficial effects on bone metabolism. Polysaccharide molecules often have difficulty passing through the intestinal cell membrane and are directly absorbed in the gastrointestinal tract. Therefore, polysaccharides may affect intestinal flora and play a role in disease treatment. We performed a comprehensive review of the relevant literature published from 2003 to 2023. We found that several polysaccharides from traditional Chinese medicines, including Astragalus, Achyranthes bidentata and Eucommia ulmoides, and the polysaccharides from several dietary fibers mainly composed of inulin, resistant starch, and dextran could enrich the intestinal microbiota group to regulate bone metabolism. The promotion of polysaccharide decomposition by regulating the Bacteroides phylum is particularly critical. Studies on the structure-activity relationship showed that molecular weight, glycosidic bonds, and monosaccharide composition may affect the ability of polysaccharides. The mechanism by which polysaccharides regulate intestinal flora to enhance bone metabolism may be related to the regulation of short-chain fatty acids, immunity, and hormones, involving some signaling pathways, such as TGF-β, Wnt/β-catenin, BMP/Smads, and RANKL. This paper provides a useful reference for the study of polysaccharides and suggests their potential application in the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Yun Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Yun Jie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Cheng Yan Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Li Zou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chao Ying Tong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China; College of Chemistry and Chemical Engineering,Central South University, Changsha, Hunan 410083, PR China
| | - Yang Zhang
- Institute of Orthopedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Dan Shou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
25
|
Capeding MRZ, Phee LCM, Ming C, Noti M, Vidal K, Le Carrou G, Frézal A, Moll JM, Vogt JK, Myers PN, Nielsen BH, Boulangé CL, Samuel TM, Berger B, Cercamondi CI. Safety, efficacy, and impact on gut microbial ecology of a Bifidobacterium longum subspecies infantis LMG11588 supplementation in healthy term infants: a randomized, double-blind, controlled trial in the Philippines. Front Nutr 2023; 10:1319873. [PMID: 38162520 PMCID: PMC10755859 DOI: 10.3389/fnut.2023.1319873] [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: 10/17/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Bifidobacterium longum subspecies infantis (B. infantis) may play a key role in infant gut development. This trial evaluated safety, tolerability, and efficacy of B. infantis LMG11588 supplementation. Methods This randomized, placebo-controlled, double-blind study conducted in the Philippines included healthy breastfed and/or formula-fed infants (14-21 days old) randomized for 8 weeks to a control group (CG; n = 77), or any of two B. infantis experimental groups (EGs): low (Lo-EG; 1*108 CFU/day; n = 75) or high dose (Hi-EG; 1.8*1010 CFU/day; n = 76). Primary endpoint was weight gain; secondary endpoints included stooling patterns, gastrointestinal symptoms, adverse events, fecal microbiome, biomarkers, pH, and organic acids. Results Non-inferiority in weight gain was demonstrated for Hi-EG and Lo-EG vs. CG. Overall, probiotic supplementation promoted mushy-soft stools, fewer regurgitation episodes, and increased fecal acetate production, which was more pronounced in the exclusively breastfed infants (EBF) and positively correlated with B. infantis abundance. In EBF, fecal pro-inflammatory cytokines (IL-1 beta, IL-8) were reduced. Strain-level metagenomic analysis allowed attributing the increased abundance of B. infantis in EGs versus CG, to LMG11588 probiotic colonization. Colonization by autochthonous B. infantis strains was similar between groups. Discussion B. infantis LMG11588 supplementation was associated with normal infant growth, was safe and well-tolerated and promoted a Bifidobacterium-rich microbiota driven by B. infantis LMG11588 colonization without disturbing the natural dispersal of autochthonous B. infantis strains. In EBF, supplementation stimulated microbial metabolic activity and beneficially modulated enteric inflammation.
Collapse
Affiliation(s)
| | | | - Chang Ming
- Biostatistics & Data, Nestlé Research, Lausanne, Switzerland
| | - Mario Noti
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Karine Vidal
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Gilles Le Carrou
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - A. Frézal
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | | | | | | | - Claire L. Boulangé
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Tinu Mary Samuel
- Nestlé Product Technology Center – Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| | - Bernard Berger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Colin Ivano Cercamondi
- Nestlé Product Technology Center – Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| |
Collapse
|
26
|
Batta VK, Rao SC, Patole SK. Bifidobacterium infantis as a probiotic in preterm infants: a systematic review and meta-analysis. Pediatr Res 2023; 94:1887-1905. [PMID: 37460707 PMCID: PMC10665187 DOI: 10.1038/s41390-023-02716-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/08/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Bifidobacterium infantis has special abilities to utilise human milk oligosaccharides. Hence we hypothesised that probiotic supplements containing B. infantis may confer greater benefits to preterm infants than probiotic supplements without B. infantis. METHODS A systematic review with meta-analysis was conducted according to standard guidelines. We selected RCTs evaluating probiotics compared to placebo or no treatment in preterm and/or low birth weight infants. Probiotic effects on Necrotizing Enterocolitis (NEC), Late Onset Sepsis (LOS) and Mortality were analysed separately for RCTs in which the supplemented probiotic product contained B. infantis and those that did not contain B. infantis. RESULTS 67 RCTs were included (n = 14,606), of which 16 used probiotics containing B. infantis (Subgroup A) and 51 RCTs did not (Subgroup B) Meta-analysis of all RCTs indicated that probiotics reduced the risk of NEC, LOS, and mortality. The subgroup meta-analysis demonstrated greater reduction in the incidence of NEC in subgroup A than subgroup B [(relative risk in subgroup A: 0.38; 95% CI, 0.27-0.55) versus (0.67; 95% CI, 0.55-0.81) in subgroup B; p value for subgroup difference: 0.01]. CONCLUSIONS These results provide indirect evidence that probiotic supplements that include B. infantis may be more beneficial for preterm infants. Well-designed RCTs are necessary to confirm these findings. IMPACT Evidence is emerging that beneficial effects of probiotics are species and strain specific. This systematic review analyses if B. infantis supplementation provides an advantage to preterm infants. This is the first systematic review evaluating the effects of probiotics containing B. infantis in preterm infants. The results of this systematic review provides indirect evidence that probiotics that include B. infantis may be more beneficial for preterm infants. These results will help in guiding future research and clinical practice for using B. infantis as a probiotic in preterm infants.
Collapse
Affiliation(s)
- Vamsi K Batta
- Neonatal Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia
- Neonatal Intensive Care Unit, King Edward Memorial Hospital, Perth, WA, Australia
| | - Shripada C Rao
- Neonatal Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia.
- School of Medicine, University of Western Australia, Perth, WA, Australia.
| | - Sanjay K Patole
- Neonatal Intensive Care Unit, King Edward Memorial Hospital, Perth, WA, Australia
- School of Medicine, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
27
|
Chen Y, Lu Y, Wang T, Wu J, Yu B. Changes in Gut Microbiota at 1-60 Days in 92 Preterm Infants in a Neonatal Intensive Care Unit Using 16S rRNA Gene Sequencing. Med Sci Monit 2023; 29:e941560. [PMID: 38018034 DOI: 10.12659/msm.941560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Neonatal gut diversity is influenced by birth conditions and probiotic/antibiotic use. The gut microbiota affects brain development, immunity, and risk of diseases. Preterm infants, especially in neonatal intensive care units (NICUs), have different gut flora from full-term infants, suggesting in utero microbial colonization. This study examined gut microbiota changes in 92 NICU preterm infants in China. MATERIAL AND METHODS We collected data on 92 preterm infants admitted to the NICU immediately after birth, and fecal samples were collected on days 1, 3, 7, 14, 21, 28, and 60. We analyzed changes in intestinal bacteria through 16S rRNA sequencing, predicted the change in gut microbiota function over time, and compared the effects of main feeding modality on the intestinal bacteria of preterm infants. RESULTS At the phylum level, the top 5 phyla in total accounted for 99.69% of the abundance, in decreasing order of abundance: Proteobacteria, Firmicutes, Actinobacteria, Tenericutes, and Bacteroidetes. At the genus level, the top 10 genera in terms of abundance accounted for a total of 90.90%, in decreasing order of abundance: Pseudomonas, Staphylococcus, Klebsiella, Escherichia-Shigella, unclassified Enterobacteriaceae, Staphylococcus, Clostridium-sensu-stricto-1, Streptococcus, Sphingomonas, and Ureaplasma. The abundance of Proteobacteria and Pseudomonas showed a decreasing trend at first, reached a minimum at day 14, and then an increasing trend, while the opposite trend was observed for Firmicutes. The metabolic function of the bacterial community changed greatly at different time points. The abundance of Proteobacteria at the phylum level and Streptococcus at the genus level in formula-fed infants were significantly higher than in breast-fed infants. CONCLUSIONS Between 1 and 60 days, the gut microbiome in preterm infants in the NICU changed with changes in feeding patterns, with the main gut bacteria being from the phyla, Proteobacteria, and Pseudomonas.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China (mainland)
| | - Yanbo Lu
- Ningbo Women's and Children's Hospital, Ningbo, Zhejiang, China (mainland)
| | - Ting Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China (mainland)
| | - Junhua Wu
- Ningbo Women's and Children's Hospital, Ningbo, Zhejiang, China (mainland)
| | - Beirong Yu
- Ningbo Women's and Children's Hospital, Ningbo, Zhejiang, China (mainland)
| |
Collapse
|
28
|
Lou YC, Rubin BE, Schoelmerich MC, DiMarco KS, Borges AL, Rovinsky R, Song L, Doudna JA, Banfield JF. Infant microbiome cultivation and metagenomic analysis reveal Bifidobacterium 2'-fucosyllactose utilization can be facilitated by coexisting species. Nat Commun 2023; 14:7417. [PMID: 37973815 PMCID: PMC10654741 DOI: 10.1038/s41467-023-43279-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
The early-life gut microbiome development has long-term health impacts and can be influenced by factors such as infant diet. Human milk oligosaccharides (HMOs), an essential component of breast milk that can only be metabolized by some beneficial gut microorganisms, ensure proper gut microbiome establishment and infant development. However, how HMOs are metabolized by gut microbiomes is not fully elucidated. Isolate studies have revealed the genetic basis for HMO metabolism, but they exclude the possibility of HMO assimilation via synergistic interactions involving multiple organisms. Here, we investigate microbiome responses to 2'-fucosyllactose (2'FL), a prevalent HMO and a common infant formula additive, by establishing individualized microbiomes using fecal samples from three infants as the inocula. Bifidobacterium breve, a prominent member of infant microbiomes, typically cannot metabolize 2'FL. Using metagenomic data, we predict that extracellular fucosidases encoded by co-existing members such as Ruminococcus gnavus initiate 2'FL breakdown, thus critical for B. breve's growth. Using both targeted co-cultures and by supplementation of R. gnavus into one microbiome, we show that R. gnavus can promote extensive growth of B. breve through the release of lactose from 2'FL. Overall, microbiome cultivation combined with genome-resolved metagenomics demonstrates that HMO utilization can vary with an individual's microbiome.
Collapse
Affiliation(s)
- Yue Clare Lou
- Department of Plant and Microbial Biology, University of California, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Benjamin E Rubin
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Marie C Schoelmerich
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Environmental Systems Sciences, ETH Zurich, Zurich, Switzerland
| | - Kaden S DiMarco
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Adair L Borges
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Rachel Rovinsky
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Leo Song
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Jennifer A Doudna
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Jillian F Banfield
- Innovative Genomics Institute, University of California, Berkeley, CA, USA.
- Department of Earth and Planetary Science, University of California, Berkeley, CA, USA.
- Department of Environmental Science, Policy, and Management, University of California, Berkeley, CA, USA.
| |
Collapse
|
29
|
Wang K, Xin Z, Chen Z, Li H, Wang D, Yuan Y. Progress of Conjugated Linoleic Acid on Milk Fat Metabolism in Ruminants and Humans. Animals (Basel) 2023; 13:3429. [PMID: 37958184 PMCID: PMC10647460 DOI: 10.3390/ani13213429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
As a valuable nutrient in milk, fat accounts for a significant proportion of the energy requirements of ruminants and is largely responsible for determining milk quality. Fatty acids (FAs) are a pivotal component of milk fat. Conjugated linoleic acid (CLA) is one of the naturally occurring FAs prevalent in ruminant dairy products and meat. Increasing attention has been given to CLA because of its anti-cancer, anti-inflammatory, immune regulation, and lipid metabolism regulation properties, and these benefits potentially contribute to the growth and health of infants. In breast milk, CLA is present in trace amounts, mainly in the form of cis-9, trans-11 CLA. Notably, cis-9, trans-11 CLA improves the milk fat rate while trans-10, cis-12 CLA inhibits it. Apart from having multiple physiological functions, CLA is also a pivotal factor in determining the milk quality of ruminants, especially milk fat rate. In response to growing interest in green and healthy functional foods, more and more researchers are exploring the potential of CLA to improve the production performance of animals and the nutritional value of livestock products. Taken together, it is novel and worthwhile to investigate how CLA regulates milk fat synthesis. It is the purpose of this review to clarify the necessity for studying CLA in ruminant milk fat and breast milk fat.
Collapse
Affiliation(s)
- Kun Wang
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou 310058, China; (K.W.); (Z.X.)
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China;
| | - Zimeng Xin
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou 310058, China; (K.W.); (Z.X.)
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China;
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China;
| | - Huanan Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China;
| | - Diming Wang
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou 310058, China; (K.W.); (Z.X.)
| | - Yuan Yuan
- School of Nursing, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
30
|
Urakami C, Yamanouchi S, Kimata T, Tsuji S, Akagawa S, Kino J, Akagawa Y, Kato S, Araki A, Kaneko K. Abnormal Development of Microbiota May Be a Risk Factor for Febrile Urinary Tract Infection in Infancy. Microorganisms 2023; 11:2574. [PMID: 37894232 PMCID: PMC10609410 DOI: 10.3390/microorganisms11102574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Febrile urinary tract infection (fUTI) is common in infants, but specific risk factors for developing it remain unclear. As most fUTIs are caused by ascending infections of intestinal bacteria, dysbiosis-an imbalance in gut microbial communities-may increase fUTI risk. This study was conducted to test the hypothesis that abnormal development of gut microbiota during infancy increases the risk of developing fUTI. Stool samples were collected from 28 infants aged 3-11 months with first-onset fUTI (fUTI group) and 51 healthy infants of the same age (HC group). After bacterial DNA extraction, 16S rRNA expression was measured and the diversity of gut microbiota and constituent bacteria were compared between the two groups. The alpha diversity of gut microbiota (median Shannon index and Chao index) was significantly lower in the fUTI group (3.0 and 42.5) than in the HC group (3.7 and 97.0; p < 0.001). The beta diversity also formed different clusters between the two groups (p < 0.001), suggesting differences in their microbial composition. The linear discriminant analysis effect size showed that the fUTI group proportionally featured significantly more Escherichia-Shigella in the gut microbiota (9.5%) than the HC group (3.1%; p < 0.001). In summary, abnormal gut microbiota development during infancy may increase the risk of fUTI.
Collapse
Affiliation(s)
- Chika Urakami
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
- Osaka Asahi Children’s Hospital, Osaka 535-0022, Japan; (J.K.); (A.A.)
| | - Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Takahisa Kimata
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Jiro Kino
- Osaka Asahi Children’s Hospital, Osaka 535-0022, Japan; (J.K.); (A.A.)
| | - Yuko Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Shogo Kato
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| | - Atsushi Araki
- Osaka Asahi Children’s Hospital, Osaka 535-0022, Japan; (J.K.); (A.A.)
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan; (C.U.); (S.Y.); (T.K.); (S.T.); (S.A.); (Y.A.); (S.K.)
| |
Collapse
|
31
|
Vogel K, Arra A, Lingel H, Bretschneider D, Prätsch F, Schanze D, Zenker M, Balk S, Bruder D, Geffers R, Hachenberg T, Arens C, Brunner-Weinzierl MC. Bifidobacteria shape antimicrobial T-helper cell responses during infancy and adulthood. Nat Commun 2023; 14:5943. [PMID: 37741816 PMCID: PMC10517955 DOI: 10.1038/s41467-023-41630-x] [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: 12/09/2022] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
Microbial infections early in life are challenging for the unexperienced immune system. The SARS-CoV-2 pandemic again has highlighted that neonatal, infant, child, and adult T-helper(Th)-cells respond differently to infections, and requires further understanding. This study investigates anti-bacterial T-cell responses against Staphylococcus aureus aureus, Staphylococcus epidermidis and Bifidobacterium longum infantis in early stages of life and adults and shows age and pathogen-dependent mechanisms. Beside activation-induced clustering, T-cells stimulated with Staphylococci become Th1-type cells; however, this differentiation is mitigated in Bifidobacterium-stimulated T-cells. Strikingly, prestimulation of T-cells with Bifidobacterium suppresses the activation of Staphylococcus-specific T-helper cells in a cell-cell dependent manner by inducing FoxP3+CD4+ T-cells, increasing IL-10 and galectin-1 secretion and showing a CTLA-4-dependent inhibitory capacity. Furthermore Bifidobacterium dampens Th responses of severely ill COVID-19 patients likely contributing to resolution of harmful overreactions of the immune system. Targeted, age-specific interventions may enhance infection defence, and specific immune features may have potential cross-age utilization.
Collapse
Affiliation(s)
- Katrin Vogel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Florian Prätsch
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Denny Schanze
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Silke Balk
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Hachenberg
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christoph Arens
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
- Justus-Liebig-University Gießen, University Hospital of Gießen and Marburg (UKGM), Gießen Campus, Department of Otorhinolaryngology, Head/Neck Surgery and Plastic Surgery, Gießen, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
32
|
Murakami R, Yoshida K, Sakanaka M, Urashima T, Xiao JZ, Katayama T, Odamaki T. Preferential sugar utilization by bifidobacterial species. MICROBIOME RESEARCH REPORTS 2023; 2:31. [PMID: 38045925 PMCID: PMC10688810 DOI: 10.20517/mrr.2023.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/28/2023] [Accepted: 08/16/2023] [Indexed: 12/05/2023]
Abstract
Aim: Bifidobacteria benefit host health and homeostasis by breaking down diet- and host-derived carbohydrates to produce organic acids in the intestine. However, the sugar utilization preference of bifidobacterial species is poorly understood. Thus, this study aimed to investigate the sugar utilization preference (i.e., glucose or lactose) of various bifidobacterial species. Methods: Strains belonging to 40 bifidobacterial species/subspecies were cultured on a modified MRS medium supplemented with glucose and/or lactose, and their preferential sugar utilization was assessed using high-performance thin-layer chromatography. Comparative genomic analysis was conducted with a focus on genes involved in lactose and glucose uptake and genes encoding for carbohydrate-active enzymes. Results: Strains that preferentially utilized glucose or lactose were identified. Almost all the lactose-preferring strains harbored the lactose symporter lacS gene. However, the comparative genomic analysis could not explain all their differences in sugar utilization preference. Analysis based on isolate source revealed that all 10 strains isolated from humans preferentially utilized lactose, whereas all four strains isolated from insects preferentially utilized glucose. In addition, bifidobacterial species isolated from hosts whose milk contained higher lactose amounts preferentially utilized lactose. Lactose was also detected in the feces of human infants, suggesting that lactose serves as a carbon source not only for infants but also for gut microbes in vivo. Conclusion: The different sugar preference phenotypes of Bifidobacterium species may be ascribed to the residential environment affected by the dietary habits of their host. This study is the first to systematically evaluate the sugar uptake preference of various bifidobacterial species.
Collapse
Affiliation(s)
- Ryuta Murakami
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Keisuke Yoshida
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Tadasu Urashima
- Department of Food and Life Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| |
Collapse
|
33
|
Martín-Masot R, Díaz-Martín JJ, Santamaría-Orleans A, Navas-López VM. Spanish Pediatricians' Positions Regarding Prevention, Diagnosis, Nutritional Management, and Challenges in Cow's Milk Protein Allergy. Nutrients 2023; 15:3586. [PMID: 37630776 PMCID: PMC10458855 DOI: 10.3390/nu15163586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Management of cow's milk protein allergy (CMPA) can vary depending on the experience and area of expertise of the clinician responsible for the patient's follow-up, which may or may not align with the recently published literature. To analyze the perspectives of Spanish pediatricians on this topic, a survey was conducted. The survey aimed to determine the current opinions and attitudes of 222 primary care and hospital pediatricians toward CMPA prevention and nutritional management. Participating pediatricians completed the questionnaire, providing insights into their daily clinical practices, including access to testing, attitudes with respect to various aspects of CMPA diagnosis, prevention, oral food challenges, and treatment. The findings revealed that pediatricians generally agree on the use of extensively hydrolyzed formulas (eHFs) to prevent CMPA in high-risk atopic children, despite limited evidence supporting the widespread use of this practice. However, consensus was lacking regarding the utility of formulas with prebiotics and probiotics for expediting tolerance development. In most cases, pediatricians preferred eHFs for the nutritional management of CMPA, followed by hydrolyzed rice formulas (HRFs), with amino-acid-based formulas (AAFs) being the third option. Certain issues remained controversial among pediatricians, such as prevention methods, symptom assessment, and the role of probiotics. These variations in management approaches reflect the influence of clinician experience and area of expertise, underscoring the need for standardized guidelines in this field.
Collapse
Affiliation(s)
- Rafael Martín-Masot
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Málaga, 29011 Málaga, Spain (V.M.N.-L.)
| | - Juan José Díaz-Martín
- Pediatric Gastroenterology and Nutrition Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | | | - Víctor Manuel Navas-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Málaga, 29011 Málaga, Spain (V.M.N.-L.)
| |
Collapse
|
34
|
Wurm P, Stampfer L, Greimel T, Leitner E, Zechner EL, Bauchinger S, Hauer AC, Gorkiewicz G, Högenauer C, Hoffmann KM. Gut Microbiota Dysbiosis in Suspected Food Protein Induced Proctocolitis-A Prospective Comparative Cohort Trial. J Pediatr Gastroenterol Nutr 2023; 77:31-38. [PMID: 37040073 DOI: 10.1097/mpg.0000000000003789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
OBJECTIVES In infants with suspected food protein induced proctocolitis (sFPIP) only a minority of patients are finally diagnosed with the disease following diagnostic dietary intervention (DDI). There is a need for a pathophysiological explanation for the cause of hematochezia in the majority of sFPIP infants. METHODS We prospectively recruited infants with sFPIP and healthy controls. Fecal samples were collected at inclusion, week 4 (end of DDI in sFPIP), and week 8. For 16S rRNA sequencing (515F/806R) we used Illumina MiSeq sequencing system. Amplicon sequence variants were generated using Qiime2 and DADA2. Qiime diversity alpha and beta group comparisons and linear discriminant analysis effect size analysis was performed. For shotgun metagenomic analysis on species level we used KneadData and MetaPhlAn2. RESULTS Fourteen sFPIP infants were compared to 55 healthy infants. At inclusion overall microbial composition of sFPIP infants differed significantly from controls (weighted UniFrac; Pairwise PERMANOVA, P = 0.002, pseudo- F = 5.008). On genus level healthy infant microbiota was significantly enriched with Bifidobacterium ( B ) compared to sFPIP patients (linear discriminant analysis [LDA] = 5.5, P < 0.001, 31.3% vs 12.1%). sFPIP stool was significantly enriched by Clostridium sensu stricto 1 over controls (LDA = 5.3, P = 0.003, 3.5% vs 18.3%). DDI caused a significant and sustained increase of Bifidobacterium (LDA = 5.4, P = 0.048, 27.9%) in sFPIP infants. Species level analysis revealed significant reduction of abundance of B longum in sFPIP patients, which after DDI was reversed by B. species other than B longum . CONCLUSIONS We revealed a gut microbiota dysbiosis phenomenon in sFPIP infants. DDI induces a microbiota composition comparable to that of healthy infants. In most sFPIP infants hematochezia might be triggered by a gut microbiota dysbiosis phenomenon.
Collapse
Affiliation(s)
- Philipp Wurm
- From Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Laura Stampfer
- the Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Theresa Greimel
- the Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Eva Leitner
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - Sebastian Bauchinger
- the Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Almuthe C Hauer
- the Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Gregor Gorkiewicz
- From Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - K Martin Hoffmann
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
- Kinderärzte Zentrum Graz-Raaba, Raaba-Grambach, Austria
| |
Collapse
|
35
|
Krupa-Kotara K, Grajek M, Grot M, Czarnota M, Wypych-Ślusarska A, Oleksiuk K, Głogowska-Ligus J, Słowiński J. Pre- and Postnatal Determinants Shaping the Microbiome of the Newborn in the Opinion of Pregnant Women from Silesia (Poland). Life (Basel) 2023; 13:1383. [PMID: 37374165 PMCID: PMC10305644 DOI: 10.3390/life13061383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Pre- and postnatal factors influence the formation of the newborn's microbiome as early as birth and the intrauterine period has a substantial impact on the composition of the baby's gastrointestinal microbiota and its subsequent development. This study intends to measure pregnant women's knowledge of the importance of microbiota for the health of the newborn. The sample was selected based on defined inclusion and exclusion criteria. The assessment of women's knowledge was assessed by the Kolmogorov-Smirnov and Kruskal-Wallis statistical tests. This study population comprised 291 adult pregnant women with a mean age of 28.4 ± 4.7 years. A total of 34% (n = 99), 35% (n = 101), and 31.3% (n = 91) were at the 1-3 trimester, respectively. The results showed that 36.4% of the women were aware that the intrauterine period changes the makeup of the gastrointestinal microbiota, whereas 5.8% exhibited awareness of the composition of the child's normal gut microbiota. Most of the women surveyed-(72.1%)-know that colonization of the tract occurs as early as the birth period. Women with student status (those who will pursue higher education in the future) and those who had given birth to the most children exhibited higher levels of knowledge.
Collapse
Affiliation(s)
- Karolina Krupa-Kotara
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Mateusz Grajek
- Department of Public Health, Department of Public Health Policy, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland;
| | - Martina Grot
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
- Doctoral School, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Martina Czarnota
- Student Scientific Society, Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (M.G.); (M.C.)
| | - Agata Wypych-Ślusarska
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Klaudia Oleksiuk
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Joanna Głogowska-Ligus
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| | - Jerzy Słowiński
- Department of Epidemiology, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 41-902 Bytom, Poland; (A.W.-Ś.); (K.O.); (J.G.-L.); (J.S.)
| |
Collapse
|
36
|
Chichlowski M, van Diepen JA, Prodan A, Olga L, Ong KK, Kortman GAM, Dunger DB, Gross G. Early development of infant gut microbiota in relation to breastfeeding and human milk oligosaccharides. Front Nutr 2023; 10:1003032. [PMID: 36969811 PMCID: PMC10034312 DOI: 10.3389/fnut.2023.1003032] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/09/2023] [Indexed: 03/11/2023] Open
Abstract
BackgroundInfant gut microbiota composition is influenced by various factors early in life. Here, we investigate associations between infant gut microbiome development, infant age, breastfeeding duration, and human milk oligosaccharides (HMO) composition in breastmilk.MethodsA total of 94 mother-infant pairs were recruited as part of the Cambridge Baby Growth and Breastfeeding Study (CBGS-BF) (Cambridge, UK). Infant stool samples (n = 337) were collected at 2 week, 6 week, 3 month, and 6 month of age. The 16S rRNA V3-V4 rRNA region was sequenced using MiSeq Illumina to determine microbiota composition and diversity. Mother’s hindmilk samples were collected at birth, 2 week, 6 week, 3 month, and 6 month postpartum. Concentrations of five neutral [2′FL, 3′FL, lacto-N-fucopentaose 1 (LNFP1), LNnT, LNT] and two acidic (3′SL, and 6′SL) HMOs were measured in all milk samples using High-Performance Anion-Exchange Chromatography with Pulsed Amperometric Detection (HPAEC-PAD). We explored the associations between infant gut microbiome parameters and age, duration of exclusive breastfeeding (EBF), and levels of individual HMOs.ResultsBifidobacterium was the most abundant genus in infant stool at all-time points, irrespective of breastfeeding duration, with an overall mean relative abundance of 70%. The relative abundance of B. bifidum in stool from infants who were breastfed for longer than 6 months was significantly higher compared to the infant breastfed up to 3 months (p = 0.0285). Alpha-diversity (both Shannon and ASV-level Richness) of infant gut microbiota showed a biphasic change with infant age, decreasing from 2 weeks until 3 months and then increasing until 6 months of age. Bifidobacterium relative abundance was associated with higher concentrations of 2′FL and LNFP1 in breastmilk across all time-points (p = 0.049 and 0.017, respectively), with trends toward a higher abundance of B. longum species. No significant association with Bifidobacterium was found for breastmilk LNnT, 3′SL, and 6′SL levels.ConclusionOur study is in line with previous data demonstrating that EBF duration in the first months of life impacts infant gut microbiota composition. The observed links between specific HMOs in breastmilk and bacteria in infant stool provide evidence of how mother’s milk affects infant microbiome development.
Collapse
Affiliation(s)
- Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
| | - Janna A. van Diepen
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Nijmegen, Netherlands
- *Correspondence: Janna A. van Diepen,
| | | | - Laurentya Olga
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Ken K. Ong
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | | | - David B. Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Nijmegen, Netherlands
| |
Collapse
|
37
|
A Scoping Review of Neonatal Opioid Withdrawal and the Infant Gut Microbiome: Does Human Milk Optimize Infant Outcomes? Adv Neonatal Care 2023; 23:237-245. [PMID: 36867674 DOI: 10.1097/anc.0000000000001056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
BACKGROUND While a growing body of literature has established the role of human milk as a mechanism of protection in the formation of the infant gut microbiome, it remains unclear the extent to which this association exists for infants with neonatal opioid withdrawal syndrome. PURPOSE The purpose of this scoping review was to describe the current state of the literature regarding the influence of human milk on infant gut microbiota in infants with neonatal opioid withdrawal syndrome. DATA SOURCES CINAHL, PubMed, and Scopus databases were searched for original studies published from January 2009 through February 2022. Additionally, unpublished studies across relevant trial registries, conference proceedings, websites, and organizations were reviewed for possible inclusion. A total of 1610 articles met selection criteria through database and register searches and 20 through manual reference searches. STUDY SELECTION Inclusion criteria were primary research studies, written in English, published between 2009 and 2022, including a sample of infants with neonatal opioid withdrawal syndrome/neonatal abstinence syndrome, and focusing on the relationship between the receipt of human milk and the infant gut microbiome. DATA EXTRACTION Two authors independently conducted title/abstract and full-text review until there was consensus of study selection. RESULTS No studies satisfied the inclusion criteria, which resulted in an empty review. IMPLICATIONS FOR PRACTICE AND RESEARCH Findings from this study document the paucity of data exploring the associations between human milk, the infant gut microbiome, and subsequent neonatal opioid withdrawal syndrome. Further, these results highlight the timely importance of prioritizing this area of scientific inquiry.
Collapse
|
38
|
Breastfeeding enrichment of B. longum subsp. infantis mitigates the effect of antibiotics on the microbiota and childhood asthma risk. MED 2023; 4:92-112.e5. [PMID: 36603585 DOI: 10.1016/j.medj.2022.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/09/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Early antibiotic exposure is linked to persistent disruption of the infant gut microbiome and subsequent elevated pediatric asthma risk. Breastfeeding acts as a primary modulator of the gut microbiome during early life, but its effect on asthma development has remained unclear. METHODS We harnessed the CHILD cohort to interrogate the influence of breastfeeding on antibiotic-associated asthma risk in a subset of children (n = 2,521). We then profiled the infant microbiomes in a subset of these children (n = 1,338) using shotgun metagenomic sequencing and compared human milk oligosaccharide and fatty acid composition from paired maternal human milk samples for 561 of these infants. FINDINGS Children who took antibiotics without breastfeeding had 3-fold higher asthma odds, whereas there was no such association in children who received antibiotics while breastfeeding. This benefit was associated with widespread "re-balancing" of taxonomic and functional components of the infant microbiome. Functional changes associated with asthma protection were linked to enriched Bifidobacterium longum subsp. infantis colonization. Network analysis identified a selection of fucosylated human milk oligosaccharides in paired maternal samples that were positively associated with B. infantis and these broader functional changes. CONCLUSIONS Our data suggest that breastfeeding and antibiotics have opposing effects on the infant microbiome and that breastfeeding enrichment of B. infantis is associated with reduced antibiotic-associated asthma risk. FUNDING This work was supported in part by the Canadian Institutes of Health Research; the Allergy, Genes and Environment Network of Centres of Excellence; Genome Canada; and Genome British Columbia.
Collapse
|
39
|
Rahman MS, Lee Y, Park DS, Kim YS. Bifidobacterium bifidum DS0908 and Bifidobacterium longum DS0950 Culture-Supernatants Ameliorate Obesity-Related Characteristics in Mice with High-Fat Diet-Induced Obesity. J Microbiol Biotechnol 2023; 33:96-105. [PMID: 36457182 PMCID: PMC9899789 DOI: 10.4014/jmb.2210.10046] [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: 10/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
Probiotic supplements have promising therapeutic effects on chronic diseases. In this study, we demonstrated the anti-obesity effects of two potential probiotics, Bifidobacterium bifidum DS0908 (DS0908) and Bifidobacterium longum DS0950 (DS0950). Treatment with DS0908 and DS0950 postbiotics significantly induced the expression of the brown adipocyte-specific markers UCP1, PPARγ, PGC1α, PRDM16 and beige adipocyte-specific markers CD137, FGF21, P2RX5, and COX2 in C3H10T1/2 mesenchymal stem cells (MSCs). In mice with high-fat diet (HFD)-induced obesity, both potential probiotics and postbiotics noticeably reduced body weight and epididymal fat accumulation without affecting food intake. DS0908 and DS0950 also improved insulin sensitivity and glucose use in mice with HFD-induced obesity. In addition, DS0908 and DS0950 improved the plasma lipid profile, proved by reduced triglyceride, low-density lipoprotein, and cholesterol levels. Furthermore, DS0908 and DS0950 improved mitochondrial respiratory function, confirmed by the high expression of oxidative phosphorylation proteins, during thermogenesis induction in the visceral and epididymal fat in mice with HFD-induced obesity. Notably, the physiological and metabolic changes were more significant after treatment with potential probiotic culture-supernatants than those with the bacterial pellet. Finally, gene knockdown and co-treatment with inhibitor-mediated mechanistic analyses showed that both DS0908 and DS0950 exerted anti-obesity-related effects via the PKA/p38 MAPK signaling activation in C3H10T1/2 MSCs. Our observations suggest that DS0908 and DS0950 could potentially alleviate obesity as dietary supplements.
Collapse
Affiliation(s)
- M. Shamim Rahman
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Youri Lee
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Doo-Sang Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Yong-Sik Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Corresponding author Phone: +82-41-570-2413 Fax: +82-41-575-2412 E-mail:
| |
Collapse
|
40
|
Mills S, Yang B, Smith GJ, Stanton C, Ross RP. Efficacy of Bifidobacterium longum alone or in multi-strain probiotic formulations during early life and beyond. Gut Microbes 2023; 15:2186098. [PMID: 36896934 PMCID: PMC10012958 DOI: 10.1080/19490976.2023.2186098] [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: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
The significance of Bifidobacterium to human health can be appreciated from its early colonization of the neonatal gut, where Bifidobacterium longum represents the most abundant species. While its relative abundance declines with age, it is further reduced in several diseases. Research into the beneficial properties of B. longum has unveiled a range of mechanisms, including the production of bioactive molecules, such as short-chain fatty acids, polysaccharides, and serine protease inhibitors. From its intestinal niche, B. longum can have far-reaching effects in the body influencing immune responses in the lungs and even skin, as well as influencing brain activity. In this review, we present the biological and clinical impacts of this species on a range of human conditions beginning in neonatal life and beyond. The available scientific evidence reveals a strong rationale for continued research and further clinical trials that investigate the ability of B. longum to treat or prevent a range of diseases across the human lifespan.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Co Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
41
|
Zhou Y, Zhang F, Mao L, Feng T, Wang K, Xu M, Lv B, Wang X. Bifico relieves irritable bowel syndrome by regulating gut microbiota dysbiosis and inflammatory cytokines. Eur J Nutr 2023; 62:139-155. [PMID: 35918555 PMCID: PMC9899748 DOI: 10.1007/s00394-022-02958-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Gut microbiota dysbiosis, a core pathophysiology of irritable bowel syndrome (IBS), is closely related to immunological and metabolic functions. Gut microbiota-based therapeutics have been recently explored in several studies. Bifico is a probiotic cocktail widely used in gastrointestinal disorders which relate to the imbalance of gut microbiota. However, the efficacy and potential mechanisms of Bifico treatment in IBS remains incompletely understood. METHODS Adopting a wrap restraint stress (WRS) -induced IBS mice model. Protective effect of Bifico in IBS mice was examined through abdominal withdrawal reflex (AWR) scores. 16S rDNA, 1H nuclear magnetic resonance (1H-NMR) and western blot assays were performed to analyze alterations of gut microbiota, microbiome metabolites and inflammatory cytokines, respectively. RESULTS Bifico could decrease intestinal visceral hypersensitivity. Although gut microbiota diversity did not increase, composition of gut microbiota was changed after treatment of Bifico, which were characterized by an increase of Proteobacteria phylum and Actinobacteria phylum, Muribaculum genus, Bifidobacterium genus and a decrease of Parabacteroides genus, Sutterella genus and Lactobacillus genus. Moreover, Bifico elevated the concentration of short-chain fatty acids (SCFAs) and reduced protein levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). From further Spearman's correlation analysis, Bifidobacterium genus were positively correlated with SCFAs including propionate, butyrate, valerate and negatively correlated with IL-6 and TNF-α. CONCLUSION Bifico could alleviate symptoms of IBS mice through regulation of the gut microbiota, elevating production of SCFAs and reducing the colonic inflammatory response.
Collapse
Affiliation(s)
- Yanlin Zhou
- grid.417400.60000 0004 1799 0055Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China ,grid.268505.c0000 0000 8744 8924The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China ,Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China
| | - Fan Zhang
- grid.268505.c0000 0000 8744 8924The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China ,Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China ,grid.417400.60000 0004 1799 0055Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China
| | - Liqi Mao
- grid.411440.40000 0001 0238 8414Department of Gastroenterology, The First People’s Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, 313000 Zhejiang China
| | - Tongfei Feng
- grid.417400.60000 0004 1799 0055Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China ,Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China
| | - Kaijie Wang
- grid.417400.60000 0004 1799 0055Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China ,Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China
| | - Maosheng Xu
- grid.417400.60000 0004 1799 0055Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China
| | - Bin Lv
- grid.417400.60000 0004 1799 0055Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310003 Zhejiang China ,Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hubin Campus, Hangzhou, 310006 China
| |
Collapse
|
42
|
Chang CM, Tsai MH, Liao WC, Yang PH, Li SW, Chu SM, Huang HR, Chiang MC, Hsu JF. Effects of Probiotics on Gut Microbiomes of Extremely Preterm Infants in the Neonatal Intensive Care Unit: A Prospective Cohort Study. Nutrients 2022; 14:3239. [PMID: 35956415 PMCID: PMC9370381 DOI: 10.3390/nu14153239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 01/04/2023] Open
Abstract
Background: Probiotics have been previously reported to reduce the incidence of necrotizing enterocolitis (NEC) in extremely preterm infants, but the mechanisms by which the probiotics work remain unknown. We aimed to investigate the effects of probiotics on the gut microbiota of extremely preterm infants. Methods: A prospective cohort study was conducted on 120 extremely preterm neonates (gestational age ≤ 28 weeks) between August 2019 and December 2021. All neonates were divided into the study (receiving probiotics) and the control (no probiotics) groups. Multivariate logistic regression analysis was performed to investigate the significantly different compositions of gut microbiota between these two groups. The effects of probiotics on the occurrence of NEC and late-onset sepsis were also investigated. Results: An increased abundance of Lactobacillus was noted in neonates who received the probiotics (AOR 4.33; 95% CI, 1.89-9.96, p = 0.009) when compared with the control group. Subjects in the probiotic group had significantly fewer days of total parenteral nutrition (median [interquartile range, IQR]) 29.0 (26.8-35.0) versus 35.5 (27.8-45.0), p = 0.004) than those in the control group. The probiotic group had a significantly lower rate of late-onset sepsis than the control group (47.1% versus 70.0%, p = 0.015), but the rate of NEC, duration of hospitalization and the final in-hospital mortality rates were comparable between these two groups. Conclusions: Probiotic supplementation of extremely preterm infants soon after the initiation of feeding increased the abundance of Lactobacillus. Probiotics may reduce the risk of late-onset sepsis, but further randomized controlled trials are warranted in the future.
Collapse
Affiliation(s)
- Ching-Min Chang
- Division of Pediatric Gastrointestinal Disease, Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
| | - Ming-Horng Tsai
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology and Pediatric Hematology/Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, Yunlin 638, Taiwan
| | - Wei-Chao Liao
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 244, Taiwan
| | - Peng-Hong Yang
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology, Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Shiao-Wen Li
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 244, Taiwan
| | - Shih-Ming Chu
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 244, Taiwan
| | - Hsuan-Rong Huang
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 244, Taiwan
| | - Ming-Chou Chiang
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 244, Taiwan
| | - Jen-Fu Hsu
- College of Medicine, Chang Gung University, Taoyuan 244, Taiwan
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 244, Taiwan
| |
Collapse
|
43
|
Genomic and epigenetic landscapes drive CRISPR-based genome editing in Bifidobacterium. Proc Natl Acad Sci U S A 2022; 119:e2205068119. [PMID: 35857876 PMCID: PMC9335239 DOI: 10.1073/pnas.2205068119] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Bifidobacterium is a commensal bacterial genus ubiquitous in the human gastrointestinal tract, which is associated with a range of health benefits. The advent of CRISPR-based genome editing technologies provides opportunities to investigate the genetics of important bacteria and transcend the lack of genetic tools in bifidobacteria to study the basis for their health-promoting attributes. Here, we repurpose the endogenous type I-G CRISPR-Cas system and adopt an exogenous CRISPR base editor for genome engineering in B. animalis subsp. lactis, demonstrating that both genomic and epigenetic contexts drive editing outcomes across strains. We reprogrammed the endogenous type I-G system to screen for naturally occurring large deletions up to 27 kb and to generate a 500-bp deletion in tetW to abolish tetracycline resistance. A CRISPR-cytosine base editor was optimized to install C•G-to-T•A amber mutations to resensitize multiple B. lactis strains to tetracycline. Remarkably, we uncovered epigenetic patterns that are distributed unevenly among B. lactis strains, despite their genomic homogeneity, that may contribute to editing efficiency variability. Insights were also expanded to Bifidobacterium longum subsp. infantis to emphasize the broad relevance of these findings. This study highlights the need to develop individualized CRISPR-based genome engineering approaches for distinct bacterial strains and opens avenues for engineering of next generation probiotics.
Collapse
|
44
|
Ke A, Parreira VR, Farber JM, Goodridge L. Inhibition of Cronobacter sakazakii in an infant simulator of the human intestinal microbial ecosystem using a potential synbiotic. Front Microbiol 2022; 13:947624. [PMID: 35910651 PMCID: PMC9335077 DOI: 10.3389/fmicb.2022.947624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
Powdered infant formula (PIF) can be contaminated with Cronobacter sakazakii, which can cause severe illnesses in infants. Synbiotics, a combination of probiotics and prebiotics, could act as an alternative control measure for C. sakazakii contamination in PIF and within the infant gut, but synbiotics have not been well studied for their ability to inhibit C. sakazakii. Using a Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) inoculated with infant fecal matter, we demonstrated that a potential synbiotic, consisting of six lactic acid bacteria (LAB) strains and Vivinal GOS, can inhibit the growth of C. sakazakii in an infant possibly through either the production of antimicrobial metabolites like acetate, increasing species diversity within the SHIME compartments to compete for nutrients or a combination of mechanisms. Using a triple SHIME set-up, i.e., three identical SHIME compartments, the first SHIME (SHIME 1) was designated as the control SHIME in the absence of a treatment, whereas SHIME 2 and 3 were the treated SHIME over 2, 1-week treatment periods. The addition of the potential synbiotic (LAB + VGOS) resulted in a significant decrease in C. sakazakii levels within 1 week (p < 0.05), but in the absence of a treatment the significant decline took 2 weeks (p < 0.05), and the LAB treatment did not decrease C. sakazakii levels (p ≥ 0.05). The principal component analysis showed a distinction between metabolomic profiles for the control and LAB treatment, but similar profiles for the LAB + VGOS treatment. The addition of the potential synbiotic (LAB + VGOS) in the first treatment period slightly increased species diversity (p ≥ 0.05) compared to the control and LAB, which may have had an effect on the survival of C. sakazakii throughout the treatment period. Our results also revealed that the relative abundance of Bifidobacterium was negatively correlated with Cronobacter when no treatments were added (ρ = −0.96; p < 0.05). These findings suggest that C. sakazakii could be inhibited by the native gut microbiota, and inhibition can be accelerated by the potential synbiotic treatment.
Collapse
|
45
|
|
46
|
López-Gómez L, Antón J, López-Tofiño Y, Pomana B, Uranga JA, Abalo R. Effects of Commercial Probiotics on Colonic Sensitivity after Acute Mucosal Irritation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116485. [PMID: 35682075 PMCID: PMC9180892 DOI: 10.3390/ijerph19116485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/10/2022]
Abstract
Gastrointestinal pathologies associated with abdominal pain, such as irritable bowel syndrome or inflammatory bowel disease, lack sufficiently effective treatments. In our study we have used a rat model of visceral pain (72 animals; n = 8–13 per experimental group) to analyze the consequences of intracolonic administration of the irritant acetic acid on visceral sensitivity, histology of the colonic wall, and inflammatory response. Moreover, we have studied the possible beneficial effects of a pretreatment with a commercial probiotic (Actimel®). Contrary to expectations, acetic acid application (7 cm proximal to the anus) decreased the nociceptive response to intracolonic mechanical stimulation, with a slight increase in the histological damage of colonic mucosa. The intensity of these changes depended on the concentration (4% or 0.6%) and the time of application (30 or 60 min). Pretreatment with probiotics (by daily gavage, for 1 week) normalized the values obtained in the visceral sensitivity test but revealed an increase in the number of macrophages. These results suggest a possible activation of inhibitory mechanisms early after colonic irritation, not previously described (which need further experimental confirmation), and the ability of probiotics to normalize the effects of acetic acid. In addition, pretreatment with probiotics has a direct effect on immune functions, stimulating macrophagic activity.
Collapse
Affiliation(s)
- Laura López-Gómez
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos, 28922 Alcorcón, Spain
| | - Jaime Antón
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
| | - Yolanda López-Tofiño
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos, 28922 Alcorcón, Spain
| | - Bianca Pomana
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
| | - José A. Uranga
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos, 28922 Alcorcón, Spain
- Correspondence: (J.A.U.); (R.A.)
| | - Raquel Abalo
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (J.A.); (Y.L.-T.); (B.P.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos, 28922 Alcorcón, Spain
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
- Working Group of Basic Sciences in Pain and Analgesia of the Spanish Society of Pain, 28046 Madrid, Spain
- Correspondence: (J.A.U.); (R.A.)
| |
Collapse
|
47
|
Sharma A, Raman V, Lee J, Forbes NS. Microbial Imbalance Induces Inflammation by Promoting Salmonella Penetration through the Mucosal Barrier. ACS Infect Dis 2022; 8:969-981. [PMID: 35404574 DOI: 10.1021/acsinfecdis.1c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The balance of microbial species in the intestine must be maintained to prevent inflammation and disease. Healthy bacteria suppress infection by pathogens and prevent disorders such as inflammatory bowel diseases (IBDs). The role of mucus in the relation between pathogens and the intestinal microbiota is poorly understood. Here, we hypothesized that healthy bacteria inhibit infection by preventing pathogens from penetrating the mucus layer and that microbial imbalance leads to inflammation by promoting the penetration of the mucosal barrier. We tested this hypothesis with an in vitro model that contains mucus, an epithelial cell layer, and resident immune cells. We found that, unlike probiotic VSL#3 bacteria, Salmonella penetrated the mucosal layers and induced the production of interleukin-8 (IL-8) and tumor necrosis factor (TNF)-α. At ratios greater than 104:1, probiotic bacteria suppressed the growth and penetration of Salmonella and reduced the production of inflammatory cytokines. Counterintuitively, low densities of healthy bacteria increased both pathogen penetration and cytokine production. In all cases, mucus increased Salmonella penetration and the production of cytokines. These results suggest that mucus lessens the protective effect of probiotic bacteria by promoting barrier penetration. In this model, a more imbalanced microbial population caused infection and inflammation by selecting pathogens that are more invasive and immunogenic. Combined, the results suggest that the depletion of commensal bacteria or an insufficient dosage of probiotics could worsen an infection and cause increased inflammation. A better understanding of the interactions between pathogens, healthy microbes, and the mucosal barrier will improve the treatment of infections and inflammatory diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
48
|
Antibiotic Treatment during Pregnancy Alters Offspring Gut Microbiota in a Sex-Dependent Manner. Biomedicines 2022; 10:biomedicines10051042. [PMID: 35625778 PMCID: PMC9138679 DOI: 10.3390/biomedicines10051042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
This study investigated the effect of antibiotics administered to pregnant dams on offspring gut microbiome composition and metabolic capabilities, and how these changes in the microbiota may influence their immune responses in both the periphery and the brain. We orally administered a broad-spectrum antibiotic (ABX) cocktail consisting of vancomycin 0.5 mg/mL, ampicillin 1 mg/mL, and neomycin 1 mg/mL to pregnant dams during late gestation through birth. Bacterial DNA was extracted from offspring fecal samples, and 16S ribosomal RNA gene was sequenced by Illumina, followed by analysis of gut microbiota composition and PICRUSt prediction. Serum and brain tissue cytokine levels were analyzed by Luminex. Our results indicate that the ABX-cocktail led to significant diversity and taxonomic changes to the offspring's gut microbiome. In addition, the predicted KEGG and MetaCyc pathways were significantly altered in the offspring. Finally, there were decreased innate inflammatory cytokines and chemokines and interleukin (IL)-17 seen in the brains of ABX-cocktail offspring in response to lipopolysaccharide (LPS) immune challenge. Our results suggest that maternal ABX can produce long-lasting effects on the gut microbiome and neuroimmune responses of offspring. These findings support the role of the early microbiome in the development of offspring gastrointestinal and immune systems.
Collapse
|
49
|
Zeng S, Wang Z, Zhang P, Yin Z, Huang X, Tang X, Shi L, Guo K, Liu T, Wang M, Qiu H. Machine learning approach identifies meconium metabolites as potential biomarkers of neonatal hyperbilirubinemia. Comput Struct Biotechnol J 2022; 20:1778-1784. [PMID: 35495115 PMCID: PMC9027383 DOI: 10.1016/j.csbj.2022.03.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
Background The gut microbiota plays an important role in the early stages of human life. Our previous study showed that the abundance of intestinal flora involved in galactose metabolism was altered and correlated with increased serum bilirubin levels in children with jaundice. We conducted the present study to systematically evaluate alterations in the meconium metabolome of neonates with jaundice and search for metabolic markers associated with neonatal jaundice. Methods We included 68 neonates with neonatal hyperbilirubinemia, also known as neonatal jaundice (NJ) and 68 matched healthy controls (HC), collected meconium samples from them at birth, and performed metabolomic analysis via liquid chromatography-mass spectrometry. Results Gut metabolites enabled clearly distinguishing the neonatal jaundice (NJ) and healthy control (HC) groups. We also identified the compositions of the gut metabolites that differed significantly between the NJ and HC groups; these differentially significant metabolites were enriched in aminyl tRNA biosynthesis; pantothenic acid and coenzyme biosynthesis; and the valine, leucine and isoleucine biosynthesis pathways. Gut branched-chain amino acid (BCAA) levels were positively correlated with serum bilirubin levels, and the area under the receiver operating characteristic curve of the random forest classifier model based on BCAAs, proline, methionine, phenylalanine and total bilirubin reached 96.9%, showing good potential for diagnostic applications. Machine learning-based causal inference analysis revealed the causal effect of BCAAs on serum total bilirubin and NJ. Conclusions Altered gut metabolites in neonates with jaundice showed that increased BCAAs and total serum bilirubin were positively correlated. BCAAs proline, methionine, phenylalanine are potential biomarkers of NJ.
Collapse
Key Words
- AUROC, the area under the ROC
- BCAA, branched-chain amino acid
- Gut microbiota
- HC, healthy controls
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LC-MS, liquid chromatography-mass spectrometry
- MSUD, maple syrup urine disease
- Machine learning
- NJ, neonatal jaundice
- OPLS-DA, orthogonal partial least squares-discriminant analysis
- PCA, the principal component analysis
- PLS, partial least-squares regression
- ROC, receiver operating characteristic
- branched-chain amino acid
- causal inference
- metabolome
- neonatal hyperbilirubinemia
Collapse
Affiliation(s)
- Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Guangdong 518109, China
| | - Peng Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Guangdong 518109, China
| | - Zhaoqing Yin
- Division of Neonatology, The People's Hospital of Dehong Autonomous Prefecture, Mangshi, Yunnan 678400, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Xisheng Tang
- Oncology Department, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Lindong Shi
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Kaiping Guo
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Ting Liu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai 201102, China
- Microbiome Therapy Center, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| |
Collapse
|
50
|
Kijner S, Cher A, Yassour M. The Infant Gut Commensal Bacteroides dorei Presents a Generalized Transcriptional Response to Various Human Milk Oligosaccharides. Front Cell Infect Microbiol 2022; 12:854122. [PMID: 35372092 PMCID: PMC8971754 DOI: 10.3389/fcimb.2022.854122] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/22/2022] [Indexed: 12/22/2022] Open
Abstract
Human milk oligosaccharides (HMOs) are a family of glycans found in breastmilk with over 200 identified structures. Despite being the third-largest solid component in breastmilk, HMOs are indigestible by infants, and they serve as food for the infant gut bacteria. Most research thus far has focused on Bifidobacterium species that harbor many glycoside hydrolases (GHs) tailored to break the carbon bonds in HMO molecules. However, there are additional microbes in the infant gut, such as Bacteroides species, with increasing evidence that they, too, are able to break-down HMOs. To study the unbiased impact of breastfeeding on the infant gut microbiome, we need to investigate the underlying mechanisms of HMO utilization by all members of the infant gut. Here, we developed an optimized system for isolating Bacteroides strains from infant stool samples. We then examined the HMO utilization capacity of multiple Bacteroides isolates by performing growth curves on six common HMOs (2'-FL, DFL, 3'-SL, 6'-SL, LNT, LNnT). Isolates often displayed similar growth characteristics on similarly-structured HMOs, like sialylated or fucosylated sugars. We identified variation in HMO utilization across multiple strains of the same species, and chose to focus here on a Bacteroides dorei isolate that was able to utilize the test HMOs. We performed RNA sequencing on B. dorei cultures, comparing the transcriptional profile in minimal media supplemented with glucose or HMOs. We showed that B. dorei employs an extensive metabolic response to HMOs. Surprisingly, there was no clear up-regulation for most GH families previously known to break-down HMOs, possibly because they were almost exclusively described in Bifidobacterium species. Instead, B. dorei exhibits a generalized response to HMOs, markedly up-regulating several shared GH families across all conditions. Within each GH family, B. dorei displays a consistent pattern of up-regulation of some genes with down-regulation of the others. This response pattern to HMOs has yet to be described in other commensals of the infant gut. Our work highlights the importance of expanding the HMO-microbiome studies beyond Bifidobacterium species, sheds light on the differences across Bacteroides strains in terms of HMO utilization, and paves the way to understanding the mechanisms enabling Bacteroides HMO utilization.
Collapse
Affiliation(s)
- Sivan Kijner
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avital Cher
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moran Yassour
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|