1
|
Akkerman R, Oerlemans MMP, Ferrari M, Fernández-Lainez C, Walvoort MTC, de Vos P. Exopolysaccharides from Bifidobacterium longum subsp. infantis and Bifidobacterium adolescentis modulate Toll-like receptor signaling. Carbohydr Polym 2025; 349:123017. [PMID: 39638524 DOI: 10.1016/j.carbpol.2024.123017] [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: 09/04/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Exopolysaccharides (EPS) from probiotic bacteria like bifidobacteria, have gained considerable attention for the beneficial effects they exert in the gastrointestinal environment. Here, we investigated whether EPS isolated from Bifidobacterium longum subsp. infantis and Bifidobacterium adolescentis can interact with Toll-like receptors (TLRs) in a structure-dependent way and subsequently we investigated whether they influence cytokine-production in dendritic cells (DCs). RESULTS EPS from both B. infantis and B. adolescentis were found to be structurally different and were able to inhibit signaling of TLR2 and TLR4 in an EPS-type dependent fashion. EPS from B. infantis was shown to have stronger inhibitory effects on TLR2/1, whereas EPS from B. adolescentis showed stronger effects for TLR2/6 and TLR4. Incubation of DCs with EPS alone had no effect, however stimulation of DCs with spend-medium of epithelial cells incubated with EPS reduced production of the cytokines MCP-1/CCL2 and TNFα. CONCLUSION Here we show that EPS from B. infantis and B. adolescentis have structure-dependent immunomodulatory effects, indicating that EPS might be important effector molecules responsible for the health benefits of bifidobacteria.
Collapse
Affiliation(s)
- Renate Akkerman
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Marjolein M P Oerlemans
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Michela Ferrari
- Department of Chemical Biology, Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Cynthia Fernández-Lainez
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands; Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Ciudad de México, Mexico
| | - Marthe T C Walvoort
- Department of Chemical Biology, Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Zhu P, Savova MV, Kindt A, Wopereis H, Belzer C, Harms AC, Hankemeier T. Exploring the Fecal Metabolome in Infants With Cow's Milk Allergy: The Distinct Impacts of Cow's Milk Protein Tolerance Acquisition and of Synbiotic Supplementation. Mol Nutr Food Res 2025; 69:e202400583. [PMID: 39665335 PMCID: PMC11704826 DOI: 10.1002/mnfr.202400583] [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: 07/26/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
SCOPE Cow's milk allergy (CMA) is one of the most prevalent food allergies in early childhood, often treated via elimination diets including standard amino acid-based formula or amino acid-based formula supplemented with synbiotics (AAF or AAF-S). This work aimed to assess the effect of cow's milk (CM) tolerance acquisition and synbiotic (inulin, oligofructose, Bifidobacterium breve M-16 V) supplementation on the fecal metabolome in infants with IgE-mediated CMA. METHODS AND RESULTS The CMA-allergic infants received AAF or AAF-S for a year during which fecal samples were collected. The samples were subjected to metabolomics analyses covering gut microbial metabolites including SCFAs, tryptophan metabolites, and bile acids (BAs). Longitudinal data analysis suggested amino acids, BAs, and branched SCFAs alterations in infants who outgrew CMA during the intervention. Synbiotic supplementation significantly modified the fecal metabolome after 6 months of intervention, including altered purine, BA, and unsaturated fatty acid levels, and increased metabolites of infant-type Bifidobacterium species: indolelactic acid and 4-hydroxyphenyllactic acid. CONCLUSION This study offers no clear conclusion on the impact of CM-tolerance acquisition on the fecal metabolome. However, our results show that 6 months of synbiotic supplementation successfully altered fecal metabolome and suggest induced bifidobacteria activity, which subsequently declined after 12 months of intervention.
Collapse
Affiliation(s)
- Pingping Zhu
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Mariyana V. Savova
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Alida Kindt
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | | | - Clara Belzer
- Laboratory of MicrobiologyWageningen UniversityWageningenThe Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
3
|
Huang Y, Lu Z, Liu F, Lane JA, Chen J, Huang Q, Hu R, Zhang B. Osteopontin associated Bifidobacterium bifidum microencapsulation modulates infant fecal fermentation and gut microbiota development. Food Res Int 2024; 197:115211. [PMID: 39593296 DOI: 10.1016/j.foodres.2024.115211] [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: 06/14/2024] [Revised: 09/22/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
Probiotic supplementation is an effective method for improving infant gut health, and probiotic encapsulation can enhance probiotic viability under adverse environmental conditions while ensuring an adequate amount of probiotic is delivered to the target site to confer a health benefit for the host. In this study, Bifidobacterium bifidum R0071 was microencapsulated using pectin or alginate, combined bovine milk osteopontin (OPN) as an excipient during the microencapsulation process. The microencapsulated probiotics were subjected to in vitro simulated infant gastrointestinal digestion and a fecal fermentation model to assess survival capacity and their impact on gas and organic acid production, as well as the development of gut microbiota. The results demonstrated that microencapsulation in the presence of osteopontin increased simulated gastrointestinal survival. During infant fecal fermentation, a significant increase in total gas production (5.5-9.1 mL) was observed for the microencapsulated probiotic with even higher level of gas production observed for osteopontin associated microencapsulated probiotic during the late stage of fermentation (8-24 h). Infant fecal fermentation of the microencapsulated probiotic also produced substantial amounts of acetate (8-17 mM) and lactate (12-35 mM), along with minor amounts of succinate (1-2 mM) and propionate (0.5-2 mM). A positive correlation was observed between metabolite production and the number of viable B. bifidum R0071 entering colon fermentation, which significantly increased with the use of OPN in the microencapsulation process. The osteopontin associated microencapsulated probiotic also significantly elevated the relative abundance of Veillonella, which, along with Bifidobacterium, influenced gas and metabolite production. Overall, our findings demonstrate that incorporating OPN as an excipient in the microencapsulation of Bifidobacterium bifidum R0071 enhances probiotic viability and positively influences the development of infant gut microbiota, highlighting its potential application in promoting infant health.
Collapse
Affiliation(s)
- Yuqin Huang
- South China University of Technology, School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, Guangzhou 510640, China
| | - Zerong Lu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou 510700, China
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou 510700, China
| | - Jonathan A Lane
- H&H Group, H&H Research, Global Research and Technology Center, P61 K202 Co. Cork, Ireland
| | - Juchun Chen
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou 510700, China
| | - Qiang Huang
- South China University of Technology, School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, Guangzhou 510640, China
| | - Ruibiao Hu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou 510700, China.
| | - Bin Zhang
- South China University of Technology, School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, Guangzhou 510640, China; Sino-Singapore International Research Institute, Guangzhou 510555, China.
| |
Collapse
|
4
|
Wichmann A. Biological effects of combinations of structurally diverse human milk oligosaccharides. Front Pediatr 2024; 12:1439612. [PMID: 39564380 PMCID: PMC11573541 DOI: 10.3389/fped.2024.1439612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are a diverse group of structures and an abundant bioactive component of breastmilk that contribute to infant health and development. Preclinical studies indicate roles for HMOs in shaping the infant gut microbiota, inhibiting pathogens, modulating the immune system, and influencing cognitive development. In the past decade, several industrially produced HMOs have become available to fortify infant formula. Clinical intervention trials with manufactured HMOs have begun to corroborate some of the physiological effects reported in preclinical studies, especially modulation of the gut microbiota in the direction of breastfed infants. As more HMOs become commercially available and as HMOs have some shared mechanisms of action, there is a need to better understand the unique and differential effects of individual HMOs and the benefits of combining multiple HMOs. This review focuses on the differential effects of different HMO structural classes and individual structures and presents a scientific rationale for why combining multiple structurally diverse HMOs is expected to exert greater biological effects.
Collapse
Affiliation(s)
- Anita Wichmann
- Global Regulatory Affairs HMOs, Early Life & Medical Nutrition, DSM-Firmenich, Hørsholm, Denmark
| |
Collapse
|
5
|
Daisley BA, Allen‐Vercoe E. Microbes as medicine. Ann N Y Acad Sci 2024; 1541:63-82. [PMID: 39392836 PMCID: PMC11580781 DOI: 10.1111/nyas.15237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Over the last two decades, advancements in sequencing technologies have significantly deepened our understanding of the human microbiome's complexity, leading to increased concerns about the detrimental effects of antibiotics on these intricate microbial ecosystems. Concurrently, the rise in antimicrobial resistance has intensified the focus on how beneficial microbes can be harnessed to treat diseases and improve health and offer potentially promising alternatives to traditional antibiotic treatments. Here, we provide a comprehensive overview of both established and emerging microbe-centric therapies, from probiotics to advanced microbial ecosystem therapeutics, examine the sophisticated ways in which microbes are used medicinally, and consider their impacts on microbiome homeostasis and health outcomes through a microbial ecology lens. In addition, we explore the concept of rewilding the human microbiome by reintroducing "missing microbes" from nonindustrialized societies and personalizing microbiome modulation to fit individual microbial profiles-highlighting several promising directions for future research. Ultimately, the advancements in sequencing technologies combined with innovative microbial therapies and personalized approaches herald a new era in medicine poised to address antibiotic resistance and improve health outcomes through targeted microbiome management.
Collapse
Affiliation(s)
- Brendan A. Daisley
- Department of Molecular and Cellular BiologyUniversity of GuelphGuelphOntarioCanada
| | - Emma Allen‐Vercoe
- Department of Molecular and Cellular BiologyUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
6
|
Zhao J, Zhang G, Yang J, Qi X, Yao F, Gao Y, Li C, Liu L, Kang L. Surface proteins of Bifidobacterium bifidum DNG6 growing in 2'-fucosyllactose alleviating lipopolysaccharide-induced intestinal barrier injury in vitro. J Dairy Sci 2024; 107:8865-8873. [PMID: 38969003 DOI: 10.3168/jds.2024-25019] [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: 04/17/2024] [Accepted: 06/07/2024] [Indexed: 07/07/2024]
Abstract
Human milk oligosaccharides promote the growth and adhesion of Bifidobacteria, thus exerting multiple biological functions on intestinal epithelial cells. Bacterial surface proteins play an important role in bacterial-host intestinal epithelial interactions. In this study, we aimed to investigate the effects of surface proteins extracted from Bifidobacterium bifidum DNG6 (B. bifidum DNG6) consuming 2'-fucosyllactose (2'-FL) on Caco-2 cells monolayer barrier injury induced by lipopolysaccharide, compared with lactose and galacto-oligosaccharides. Our results indicated that 2'-FL may promote the surface proteins of B. bifidum DNG6 to improve intestinal barrier injury by positively regulating the NF-κB signaling pathway, reducing inflammation (TNF-α reduced by 50.34%, IL-6 reduced by 22.83%, IL-1β reduced by 37.91%, and IL-10 increased by 63.47%) and strengthening tight junction proteins (ZO,1 2.39×; claudin,1 2.79×; and occluding, 4.70×). The findings of this study indicate that 2'-FL can further regulate intestinal barrier damage by promoting the alteration of B. bifidum DNG6 surface proteins. The findings of this research will also provide theoretical support for the development of synbiotic formulations.
Collapse
Affiliation(s)
- Jingjing Zhao
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Guofang Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jingbo Yang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoxi Qi
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Fei Yao
- Synaura Biotechnology (Shanghai) Co., Ltd., Shanghai 200120, China
| | - Yunfeng Gao
- Heilongjiang Agricultural Products and Veterinary Medicine and Feed Technology Identification Station, 150036, China
| | - Chun Li
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| | - Libo Liu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| | - Linhui Kang
- Synaura Biotechnology (Shanghai) Co., Ltd., Shanghai 200120, China
| |
Collapse
|
7
|
Hickman B, Salonen A, Ponsero AJ, Jokela R, Kolho KL, de Vos WM, Korpela K. Gut microbiota wellbeing index predicts overall health in a cohort of 1000 infants. Nat Commun 2024; 15:8323. [PMID: 39333099 PMCID: PMC11436675 DOI: 10.1038/s41467-024-52561-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
The human gut microbiota is central in regulating all facets of host physiology, and in early life it is thought to influence the host's immune system and metabolism, affecting long-term health. However, longitudinally monitored cohorts with parallel analysis of faecal samples and health data are scarce. In our observational study we describe the gut microbiota development in the first 2 years of life and create a gut microbiota wellbeing index based on the microbiota development and health data in a cohort of nearly 1000 infants using clustering and trajectory modelling. We show that infants' gut microbiota development is highly predictable, following one of five trajectories, dependent on infant exposures, and predictive of later health outcomes. We characterise the natural healthy gut microbiota trajectory and several different dysbiotic trajectories associated with different health outcomes. Bifidobacterium and Bacteroides appear as early keystone organisms, directing microbiota development and consistently predicting positive health outcomes. A microbiota wellbeing index, based on the healthy development trajectory, is predictive of general health over the first 5 years. The results indicate that gut microbiota succession is part of infant physiological development, predictable, and malleable. This information can be utilised to improve the predictions of individual health risks.
Collapse
Affiliation(s)
- Brandon Hickman
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alise J Ponsero
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Roosa Jokela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kaija-Leena Kolho
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children's Hospital, University of Helsinki, Stenbäckinkatu 11, FI-00029, HUS, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
8
|
Noor S, Ali S, Summer M, Riaz A, Nazakat L, Aqsa. Therapeutic Role of Probiotics Against Environmental-Induced Hepatotoxicity: Mechanisms, Clinical Perspectives, Limitations, and Future. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10365-6. [PMID: 39316257 DOI: 10.1007/s12602-024-10365-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
Hepatotoxicity is one of the biggest health challenges, particularly in the context of liver diseases, often aggravated by gut microbiota dysbiosis. The gut-liver axis has been regarded as a key idea in liver health. It indicates that changes in gut flora caused by various hepatotoxicants, including alcoholism, acetaminophen, carbon tetrachloride, and thioacetamide, can affect the balance of the gut's microflora, which may lead to increased dysbiosis and intestinal permeability. As a result, bacterial endotoxins would eventually enter the bloodstream and liver, causing hepatotoxicity and inducing inflammatory reactions. Many treatments, including liver transplantation and modern drugs, can be used to address these issues. However, because of the many side effects of these approaches, scientists and medical experts are still hoping for a therapeutic approach with fewer side effects and more positive results. Thus, probiotics have become well-known as an adjunctive strategy for managing, preventing, or reducing hepatotoxicity in treating liver injury. By altering the gut microbiota, probiotics offer a secure, non-invasive, and economical way to improve liver health in the treatment of hepatotoxicity. Through various mechanisms such as regulation of gut microbiota, reduction of pathogenic overgrowth, suppression of inflammatory mediators, modification of hepatic lipid metabolism, improvement in the performance of the epithelial barrier of the gut, antioxidative effects, and modulation of mucosal immunity, probiotics play their role in the treatment and prevention of hepatotoxicity. This review highlights the mechanistic effects of probiotics in environmental toxicants-induced hepatotoxicity and current findings on this therapeutic approach's experimental and clinical trials.
Collapse
Affiliation(s)
- Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Anfah Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Laiba Nazakat
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Aqsa
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
9
|
Raval SD, Archana G. Evaluation of synbiotic combinations of commercial probiotic strains with different prebiotics in in vitro and ex vivo human gut microcosm model. Arch Microbiol 2024; 206:315. [PMID: 38904672 DOI: 10.1007/s00203-024-04030-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Exploring probiotics for their crosstalk with the host microbiome through the fermentation of non-digestible dietary fibers (prebiotics) for their potential metabolic end-products, particularly short-chain fatty acids (SCFAs), is important for understanding the endogenous host-gut microbe interaction. This study was aimed at a systematic comparison of commercially available probiotics to understand their synergistic role with specific prebiotics in SCFAs production both in vitro and in the ex vivo gut microcosm model. Probiotic strains isolated from pharmacy products including Lactobacillus sporogenes (strain not labeled), Lactobacillus rhamnosus GG (ATCC53103), Streptococcus faecalis (T-110 JPC), Bacillus mesentericus (TO-AJPC), Bacillus clausii (SIN) and Saccharomyces boulardii (CNCM I-745) were assessed for their probiotic traits including survival, antibiotic susceptibility, and antibacterial activity against pathogenic strains. Our results showed that the microorganisms under study had strain-specific abilities to persist in human gastrointestinal conditions and varied anti-infective efficacy and antibiotic susceptibility. The probiotic strains displayed variation in the utilization of six different prebiotic substrates for their growth under aerobic and anaerobic conditions. Their prebiotic scores (PS) revealed which were the most suitable prebiotic carbohydrates for the growth of each strain and suggested xylooligosaccharide (XOS) was the poorest utilized among all. HPLC analysis revealed a versatile pattern of SCFAs produced as end-products of prebiotic fermentation by the strains which was influenced by growth conditions. Selected synbiotic (prebiotic and probiotic) combinations showing high PS and high total SCFAs production were tested in an ex vivo human gut microcosm model. Interestingly, significantly higher butyrate and propionate production was found only when synbiotics were applied as against when individual probiotic or prebiotics were applied alone. qRT-PCR analysis with specific primers showed that there was a significant increase in the abundance of lactobacilli and bifidobacteria with synbiotic blends compared to pre-, or probiotics alone. In conclusion, this work presents findings to suggest prebiotic combinations with different well-established probiotic strains that may be useful for developing effective synbiotic blends.
Collapse
Affiliation(s)
- Shivani D Raval
- Department of Microbiology and Biotechnology Center, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390 002, India
| | - G Archana
- Department of Microbiology and Biotechnology Center, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390 002, India.
| |
Collapse
|
10
|
Lijing X, Mengyao Z, Jing L, Yang L, Xiaoli X. Analysis of the characteristics of intestinal microbiota after oral tolerance in infants with food protein-induced proctocolitis. Front Pediatr 2024; 12:1338294. [PMID: 38737636 PMCID: PMC11082304 DOI: 10.3389/fped.2024.1338294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Objective To understand the characteristics of the intestinal microbiota after oral tolerance in infants with food protein-induced proctocolitis (FPIAP) treated with amino acid formula and their differences from healthy children, aiming to provide a scientific basis for guiding the application of probiotics during treatment. Methods FPIAP infants were prospectively enrolled, fecal specimens were obtained, and DNA was extracted for PCR amplification of the bacterial 16S rRNA gene V4 region. Library construction and sequencing were performed, and bioinformatic analysis was performed after obtaining valid data. Results There were 36 patients in the FPIAP group: 20 males and 16 females, age 21.944 ± 13.277 months. Diarrhea with blood in the stool were the main symptom, with an average course of 14.83 ± 9.33 days. Thirty infants (83.33%) had mucus stool, 11.11% (4/36) of them experiencing vomiting, and 55.56% (20/36) of the infants displaying poor intake and weight gain, 28 (77.78%) patients with moderate eczema, 2 (5.6%) patients with chronic respiratory symptoms. The treatment time with amino acid formula was 5.51 ± 2.88 months. A control group comprising of 25 healthy infants who were full-term, natural delivery, bottle fed, and matched in terms of age (24.840 ± 12.680 months) and gender (15 males and 10 females) was selected. Anaerobic bacteria were less abundant in FPIAP infants than healthy infants (P = 4.811 × 10-5), but potentially pathogenic bacteria were more abundant (P = 0.000). The abundance of Actinobacteria was low in FPIAP infants, the abundance of Proteobacteria was high, and the abundance of Firmicutes was reduced. Bifidobacterium could be used as a bacterial genus to differentiate healthy and FPIAP infants. Both α-and β-diversity indicators of intestinal microbiota were lower in FPIAP infants. In FPIAP infants, glucose and energy metabolism and amino acid anabolism were decreased, and inflammation-related lipopolysaccharide synthesis pathways were increased. Conclusion Compared with healthy infants, FPIAP infants with oral tolerance after amino acid formula treatment had differences in the structure and diversity of intestinal microbiota, among which Bifidobacterium was significantly reduced. Trial Registration This trial was registered on https://register.clinicaltrials.gov/.
Collapse
Affiliation(s)
| | | | | | | | - Xie Xiaoli
- Department of Pediatric Gastroenterology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Alba C, Carrera M, Álvarez-Calatayud G, Arroyo R, Fernández L, Rodríguez JM. Evaluation of Safety and Beneficial Health Effects of the Human-Milk Strain Bifidobacterium breve DSM32583: An Infant Pilot Trial. Nutrients 2024; 16:1134. [PMID: 38674825 PMCID: PMC11053739 DOI: 10.3390/nu16081134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Human milk promotes the growth of bifidobacteria in the infant gut. Adding bifidobacterial species to infant formula may contribute to increasing their presence in the gut of formula-fed infants. Therefore, the safety and anti-infectious effects of Bifidobacterium breve DSM32583, a breast milk isolate, were assessed in a pilot trial involving 3-month-old infants. The infants were randomly assigned to either the probiotic (PG) or the control (CG) groups. All the infants consumed the same formula, although it was supplemented with the strain (1 × 107 cfu/g of formula) in the PG. Overall, 160 infants (80 per group) finished the intervention. Infants in CG gained more weight compared to PG (p < 0.05), but the weights for age Z-scores at 6 months were within the normal distribution for this age group. The rates of infections affecting the gastrointestinal and respiratory tracts and antibiotic therapy were significantly lower in the PG. The bifidobacterial population and the level of short-chain fatty acids were higher (p < 0.05) in the fecal samples of PG infants. No adverse events related to formula consumption were observed. In conclusion, the administration of an infant formula with B. breve DSM32583 was safe and exerted potential beneficial effects on gut health.
Collapse
Affiliation(s)
- Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Marta Carrera
- Centro de Atención Primaria Silvano, Comunidad de Madrid, 28043 Madrid, Spain;
| | | | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| |
Collapse
|
12
|
Nehmi‐Filho V, de Freitas JA, Franco LA, Martins RC, Turri JAO, Santamarina AB, Fonseca JVDS, Sabino EC, Moraes BC, Souza E, Murata GM, Costa SF, Alcântara PS, Otoch JP, Pessoa AFM. Modulation of the gut microbiome and Firmicutes phylum reduction by a nutraceutical blend in the obesity mouse model and overweight humans: A double-blind clinical trial. Food Sci Nutr 2024; 12:2436-2454. [PMID: 38628220 PMCID: PMC11016419 DOI: 10.1002/fsn3.3927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 04/19/2024] Open
Abstract
Overweight and obesity are closely linked to gut dysbiosis/dysmetabolism and disrupted De-Ritis ratio [aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio], which may contribute to chronic noncommunicable diseases onset. Concurrently, extensive research explores nutraceuticals, and health-enhancing supplements, for disease prevention or treatment. Thus, sedentary overweight volunteers were double-blind randomized into two groups: Novel Nutraceutical_(S) (without silymarin) and Novel Nutraceutical (with silymarin). Experimental formulations were orally administered twice daily over 180 consecutive days. We evaluated fecal gut microbiota, based on partial 16S rRNA sequences, biochemistry and endocrine markers, steatosis biomarker (AST/ALT ratio), and anthropometric parameters. Post-supplementation, only the Novel Nutraceutical group reduced Clostridium clostridioforme (Firmicutes), Firmicutes/Bacteroidetes ratio (F/B ratio), and De-Ritis ratio, while elevating Bacteroides caccae and Bacteroides uniformis (Bacteroidetes) in Brazilian sedentary overweight volunteers after 180 days. In summary, the results presented here allow us to suggest the gut microbiota as the action mechanism of the Novel Nutraceutical promoting metabolic hepatic recovery in obesity/overweight non-drug interventions.
Collapse
Affiliation(s)
- Victor Nehmi‐Filho
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Jessica Alves de Freitas
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Lucas Augusto Franco
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Roberta Cristina Martins
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - José Antônio Orellana Turri
- Departamento de Ginecologia e Obstetrícia, Grupo de Pesquisa em Economia da SaúdeUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Aline Boveto Santamarina
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Joyce Vanessa da Silva Fonseca
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM‐49)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Ester Cerdeira Sabino
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Bruna Carvalho Moraes
- Laboratório de Investigação Médica (LIM‐31), Laboratório Investigação Médica em Patogênese e Terapia dirigida em Onco‐Imuno‐HematologiaUniversidade de São Paulo Faculdade de Medicina, Universidade de São Paulo Hospital das ClínicasCerqueira CésarBrazil
| | | | - Gilson Masahiro Murata
- Laboratório de Investigação Médica (LIM‐29), Laboratório de Nefrologia Celular, Genética e Molecular, Departamento de Clínica MédicaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Silvia Figueiredo Costa
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM‐49)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Paulo Sérgio Alcântara
- Departamento de CirurgiaUniversidade de São Paulo Hospital Universitário de São PauloButantãBrazil
| | - José Pinhata Otoch
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
- Departamento de CirurgiaUniversidade de São Paulo Hospital Universitário de São PauloButantãBrazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
- Efeom NutritionUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| |
Collapse
|
13
|
Singh J, Sharma M, Singh H, Arora P, Utreja P, Kumar S. Formulation, Characterization and In Vitro Evaluation of Mesalamine and Bifidobacterium bifidum Loaded Hydrogel Beads in Capsule System for Colon Targeted Delivery. AAPS PharmSciTech 2024; 25:61. [PMID: 38485901 DOI: 10.1208/s12249-024-02764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/10/2024] [Indexed: 03/19/2024] Open
Abstract
Mesalamine is a first-line drug for the treatment of inflammatory bowel diseases. However, its premature release associated with marketed formulations leads to adverse effects like gastric trouble, vomiting, and diarrhoea. To minimize these side effects, colon-targeted drug delivery is essential. Besides conventional pharmacotherapy, bifidogenic probiotics with anti-inflammatory activity has been reported to elicit a significant impact on the remission of ulcerative colitis. Bifidogenic probiotics being acid-labile necessitate developing a gastro-resistant formulation for enhancing the delivery of viable cells to the colon. The present study was aimed at developing a fixed-dose unit dosage form of mucoadhesive hydrogel beads loaded with mesalamine and Bifidobacterium bifidum further encapsulated in Eudragit® capsules for the targeted drug delivery at colonic pH. The hydrogel beads were prepared by ionotropic gelation, with the effect of single and dual-crosslinking approaches on various formulation characteristics studied. Standard size 00 Eudragit® gastro-resistant capsules were prepared and the dried beads were filled inside the capsule shells. The formulation was then evaluated for various parameters, including physicochemical characterization, in vitro biocompatibility and anti-inflammatory activity. No interaction was observed between the drug and the polymers, as confirmed through FTIR, XRD, and DSC analysis. The mean particle size of the beads was ~ 457-485 µm. The optimized formulation showed a drug entrapment efficiency of 95.4 ± 2.58%. The Eudragit® capsule shells disintegrated in approximately 13 min at pH 7.4. The mucoadhesive hydrogel beads sustained the drug release above 18 h, with 50% of the drug released by the end of 12 h. The optimized formulation demonstrated significant (p < 0.05) gastro-resistance, biocompatibility, sustained drug release, cell viability, and anti-inflammatory activity.
Collapse
Affiliation(s)
- Jagtar Singh
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Near Baddowal Cantt, Ferozepur Rd, Ludhiana, Punjab, 142021, India
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Punjab, 160062, India
| | - Mohit Sharma
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Near Baddowal Cantt, Ferozepur Rd, Ludhiana, Punjab, 142021, India
| | - Harmeet Singh
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Near Baddowal Cantt, Ferozepur Rd, Ludhiana, Punjab, 142021, India
| | - Pinky Arora
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar - Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India
- Faculty of Medical Lab Sciences, PCTE Group of Institutes, Near Baddowal Cantt, Ferozepur Rd, Ludhiana, Punjab, 142021, India
| | - Puneet Utreja
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Near Baddowal Cantt, Ferozepur Rd, Ludhiana, Punjab, 142021, India
| | - Shubham Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India.
| |
Collapse
|
14
|
Lin M, Yanjun C. Research progress on the mechanism of probiotics regulating cow milk allergy in early childhood and its application in hypoallergenic infant formula. Front Nutr 2024; 11:1254979. [PMID: 38419849 PMCID: PMC10900986 DOI: 10.3389/fnut.2024.1254979] [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: 07/08/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Some infants and young children suffer from cow's milk allergy (CMA), and have always mainly used hypoallergenic infant formula as a substitute for breast milk, but some of these formulas can still cause allergic reactions. In recent years, it has been found that probiotic nutritional interventions can regulate CMA in children. Scientific and reasonable application of probiotics to hypoallergenic infant formula is the key research direction in the future. This paper discusses the mechanism and clinical symptoms of CMA in children. This review critically ex- amines the issue of how probiotics use intestinal flora as the main vector to combine with the immune system to exert physiological functions to intervene CMA in children, with a particular focus on four mechanisms: promoting the early establishment of intestinal microecological balance, regulating the body's immunity and alleviating allergic response, enhancing the intestinal mucosal barrier function, and destroying allergen epitopes. Additionally, it overviews the development process of hypoallergenic infant formula and the research progress of probiotics in hypoallergenic infant formula. The article also offers suggestions and outlines potential future research directions and ideas in this field.
Collapse
Affiliation(s)
| | - Cong Yanjun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, College of Food and Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
15
|
Rizzo SM, Vergna LM, Alessandri G, Lee C, Fontana F, Lugli GA, Carnevali L, Bianchi MG, Barbetti M, Taurino G, Sgoifo A, Bussolati O, Turroni F, van Sinderen D, Ventura M. GH136-encoding gene (perB) is involved in gut colonization and persistence by Bifidobacterium bifidum PRL2010. Microb Biotechnol 2024; 17:e14406. [PMID: 38271233 PMCID: PMC10884991 DOI: 10.1111/1751-7915.14406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Bifidobacteria are commensal microorganisms that typically inhabit the mammalian gut, including that of humans. As they may be vertically transmitted, they commonly colonize the human intestine from the very first day following birth and may persist until adulthood and old age, although generally at a reduced relative abundance and prevalence compared to infancy. The ability of bifidobacteria to persist in the human intestinal environment has been attributed to genes involved in adhesion to epithelial cells and the encoding of complex carbohydrate-degrading enzymes. Recently, a putative mucin-degrading glycosyl hydrolase belonging to the GH136 family and encoded by the perB gene has been implicated in gut persistence of certain bifidobacterial strains. In the current study, to better characterize the function of this gene, a comparative genomic analysis was performed, revealing the presence of perB homologues in just eight bifidobacterial species known to colonize the human gut, including Bifidobacterium bifidum and Bifidobacterium longum subsp. longum strains, or in non-human primates. Mucin-mediated growth and adhesion to human intestinal cells, in addition to a rodent model colonization assay, were performed using B. bifidum PRL2010 as a perB prototype and its isogenic perB-insertion mutant. These results demonstrate that perB inactivation reduces the ability of B. bifidum PRL2010 to grow on and adhere to mucin, as well as to persist in the rodent gut niche. These results corroborate the notion that the perB gene is one of the genetic determinants involved in the persistence of B. bifidum PRL2010 in the human gut.
Collapse
Affiliation(s)
- Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Laura Maria Vergna
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Ciaran Lee
- APC Microbiome Institute and School of Microbiology, Bioscience InstituteNational University of IrelandCorkIreland
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
- GenProbio srlParmaItaly
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
| | - Luca Carnevali
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Massimiliano G. Bianchi
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Margherita Barbetti
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Giuseppe Taurino
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Andrea Sgoifo
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Ovidio Bussolati
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience InstituteNational University of IrelandCorkIreland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of ParmaParmaItaly
- Interdepartmental Research Centre “Microbiome Research Hub”University of ParmaParmaItaly
| |
Collapse
|
16
|
Yang S, Cai J, Su Q, Li Q, Meng X. Human milk oligosaccharides combine with Bifidobacterium longum to form the "golden shield" of the infant intestine: metabolic strategies, health effects, and mechanisms of action. Gut Microbes 2024; 16:2430418. [PMID: 39572856 PMCID: PMC11587862 DOI: 10.1080/19490976.2024.2430418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/04/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are the third most important nutrient in human milk and are the gold standard for infant nutrition. Due to the lack of an enzyme system capable of utilizing HMOs in the infant intestine, HMOs cannot be directly utilized. Instead, they function as natural prebiotics, participating in the establishment of the intestinal microbiota as a "bifidus factor." A crucial colonizer of the early intestine is Bifidobacterium longum (B. longum), particularly its subspecies B. longum subsp. infantis, which is the most active consumer of HMOs. However, due to the structural diversity of HMOs and the specificity of B. longum strains, studies on their synergy are limited. An in-depth investigation into the mechanisms of HMO utilization by B. longum is essential for applying both as synbiotics to promote early intestinal development in infants. This review describes the colonization advantages of B. longum in the infant intestinal tract and its metabolic strategies for HMOs. It also summarizes recent studies on the effect and mechanism of B. longum and HMOs in infant intestinal development directly or indirectly through the action of metabolites. In conclusion, further structural analysis of HMOs and a deeper understanding of the interactions between B. longum and HMOs, as well as clinical trials, are necessary to lay the foundation for future practical applications as synbiotics.
Collapse
Affiliation(s)
- Shuo Yang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Junwu Cai
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qian Su
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qiaohui Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
17
|
Rahkola EN, Rautava S, Hiltunen H, Ross C, Lahti L, Isolauri E. The preterm gut microbiota and administration routes of different probiotics: a randomized controlled trial. Pediatr Res 2023; 94:1480-1487. [PMID: 37020105 PMCID: PMC10589095 DOI: 10.1038/s41390-023-02560-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Preterm children with their aberrant gut microbiota and susceptibility to infections and inflammation constitute a considerable target group for probiotic therapy to generate the age-appropriate healthy microbiota. METHODS 68 preterm neonates were randomized into five intervention groups: Beginning from the median age of 3 days, 13 children received Lactobacillus rhamnosus GG (LGG) directly orally, and 17 via the lactating mother. 14 children received LGG with Bifidobacterium lactis Bb-12 (Bb12) orally, and 10 via the lactating mother. 14 children received placebo. The children's faecal microbiota was assessed at the age of 7 days by 16S rRNA gene sequencing. RESULTS The gut microbiota compositions of the children directly receiving the probiotic combination (LGG + Bb12) were significantly different from those of the children receiving the other intervention modes or placebo (p = 0.0012; PERMANOVA), the distinction being due to an increase in the relative abundance of Bifidobacterium animalis (P < 0.00010; ANCOM-BC), and the order Lactobacillales (P = 0.020; ANCOM-BC). CONCLUSION The connection between aberrant primary gut microbiota and a heightened risk of infectious and non-communicable diseases invites effective microbiota modulation. We show that the direct, early, and brief probiotic intervention of LGG + Bb12 109 CFU each, is sufficient to modulate the gut microbiota of the preterm neonate. IMPACT Preterm children have a higher risk of several health problems partly due to their aberrant gut microbiota. More research is needed to find a safe probiotic intervention to modify the gut microbiota of preterm children. The maternal administration route via breast milk might be safer for the newborn. In our study, the early and direct administration of the probiotic combination Lactobacillus rhamnosus GG with Bifidobacterium lactis Bb-12 increased the proportion of bifidobacteria in the preterm children's gut at the age of 7 days, but the maternal administration route was not as effective.
Collapse
Affiliation(s)
- Ella-Noora Rahkola
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland.
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland.
| | - Samuli Rautava
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics, University of Helsinki and Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Henni Hiltunen
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Chandler Ross
- Department of Computing, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Erika Isolauri
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
18
|
Archer D, Perez-Muñoz ME, Tollenaar S, Veniamin S, Cheng CC, Richard C, Barreda DR, Field CJ, Walter J. The importance of the timing of microbial signals for perinatal immune system development. MICROBIOME RESEARCH REPORTS 2023; 2:11. [PMID: 38047281 PMCID: PMC10688825 DOI: 10.20517/mrr.2023.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/10/2023] [Accepted: 03/27/2023] [Indexed: 12/05/2023]
Abstract
Background: Development and maturation of the immune system begin in utero and continue throughout the neonatal period. Both the maternal and neonatal gut microbiome influence immune development, but the relative importance of the prenatal and postnatal periods is unclear. Methods: In the present study, we characterized immune cell populations in mice in which the timing of microbiome colonization was strictly controlled using gnotobiotic methodology. Results: Compared to conventional (CONV) mice, germ-free (GF) mice conventionalized at birth (EC mice) showed few differences in immune cell populations in adulthood, explaining only 2.36% of the variation in immune phenotypes. In contrast, delaying conventionalization to the fourth week of life (DC mice) affected seven splenic immune cell populations in adulthood, including dendritic cells and regulatory T cells (Tregs), explaining 29.01% of the variation in immune phenotypes. Early life treatment of DC mice with Limosilactobacillus reuteri restored splenic dendritic cells and Tregs to levels observed in EC mice, and there were strain-specific effects on splenic CD4+ T cells, CD8+ T cells, and CD11c+ F4/80+ mononuclear phagocytes. Conclusion: This work demonstrates that the early postnatal period, compared to the prenatal period, is relatively more important for microbial signals to influence immune development in mice. Our findings further show that targeted microbial treatments in early life can redress adverse effects on immune development caused by the delayed acquisition of the neonatal gut microbiome.
Collapse
Affiliation(s)
- Dale Archer
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Maria Elisa Perez-Muñoz
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Stephanie Tollenaar
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Simona Veniamin
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Christopher C. Cheng
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Center of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Caroline Richard
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Catherine J. Field
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Jens Walter
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork T12 YN60, Ireland
| |
Collapse
|
19
|
Mogmenga I, Somda MK, Ouattara CAT, Keita I, Dabiré Y, Diguță CF, Toma RC, Ezeogu LI, Ugwuanyi JO, Ouattara AS, Matei F. Promising Probiotic Properties of the Yeasts Isolated from Rabilé, a Traditionally Fermented Beer Produced in Burkina Faso. Microorganisms 2023; 11:microorganisms11030802. [PMID: 36985375 PMCID: PMC10051331 DOI: 10.3390/microorganisms11030802] [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: 02/20/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
In recent years, research on yeasts as probiotics has gained more and more interest, which will allow the development of "new" products in the probiotics market. In this context, seventeen yeast strains isolated from Rabilé, a traditional beer produced in Burkina Faso, were assessed for their probiotic attributes. The yeast identification was performed by molecular methods, including PCR-RFLP and 5.8S-ITS region sequencing. Saccharomyces cerevisiae (14 strains) was the predominantly identified species, followed by Pichia kudriavzevii (2 strains) and Rhodotorula mucilaginosa (1 strain). Except for R. mucilaginosa, all yeast strains grew well at human temperature. The yeast strains showed high resistance when they were exposed to simulated gastrointestinal conditions. Auto-aggregation ability was between 70.20 ± 10.53% and 91.82 ± 1.96%, while co-aggregation with E. coli ranged from 24.92 ± 3.96% to 80.68 ± 9.53% and with S. enterica serovar Typhimurium from 40.89 ± 8.18% to 74.06 ± 7.94%. Furthermore, the hydrophobicity of isolated strains toward n-hexane was in the range from 43.17 ± 5.07% to 70.73 ± 2.42%. All yeast strains displayed high antioxidant capabilities, and the strains did not show hemolysis halos, such that they can be considered safe. Additionally, S. cerevisiae strains strongly inhibited the growth of foodborne pathogens. This is the first preliminary study to identify and characterize the yeast strains isolated from Rabilé with interesting probiotic properties.
Collapse
Affiliation(s)
- Iliassou Mogmenga
- Laboratoire de Microbiologie et de Biotechnologies Microbiennes, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
- Centre Universitaire de Banfora, Université Nazi BONI, Bobo-Dioulasso 01 BP 1091, Burkina Faso
| | - Marius Kounbèsiounè Somda
- Laboratoire de Microbiologie et de Biotechnologies Microbiennes, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Cheik Amadou Tidiane Ouattara
- Laboratoire de Microbiologie et de Biotechnologies Microbiennes, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Ibrahim Keita
- Laboratoire de Microbiologie et de Biotechnologies Microbiennes, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Yérobessor Dabiré
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
- Laboratoire de Biochimie, Biotechnologie, Technologie Alimentaire et Nutrition (LABIOTAN), Département de Biochimie Microbiologie, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Camelia Filofteia Diguță
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania
| | - Radu Cristian Toma
- Laboratoire de Biochimie, Biotechnologie, Technologie Alimentaire et Nutrition (LABIOTAN), Département de Biochimie Microbiologie, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Lewis I Ezeogu
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
| | - Jerry O Ugwuanyi
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
| | - Aboubakar S Ouattara
- Laboratoire de Microbiologie et de Biotechnologies Microbiennes, Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso
| | - Florentina Matei
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania
| |
Collapse
|
20
|
Camargo ARO, Van Mastrigt O, Bongers RS, Ben-Amor K, Knol J, Smid EJ, Abee T. Enhanced stress resistance of Bifidobacterium breve NRBB57 by induction of stress proteins at near-zero growth rates. Benef Microbes 2023; 14:85-94. [PMID: 36790092 DOI: 10.3920/bm2022.0074] [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: 02/16/2023]
Abstract
Bifidobacterium breve is a common habitant of the human gut and is used as probiotic in functional foods. B. breve has to cope with multiple stress conditions encountered during processing and passage through the human gut, including high temperature, low pH and exposure to oxygen. Additionally, during industrial processing and in the gut, B. breve could encounter nutrient limitation resulting in reduced growth rates that can trigger adaptive stress responses. For this reason, it is important to develop culture methods that elicit resistance to multiple stresses (robustness) encountered by the bacteria. To investigate the impact of caloric restriction on robustness of the probiotic B. breve NRBB57, this strain was grown in lactose-limited chemostat cultures and in retentostat for 21 days, at growth rates ranging from 0.4 h-1 to 0.00081 h-1. Proteomes of cells harvested at different growth rates were correlated to acid, hydrogen peroxide and heat stress survival capacity. Comparative proteome analysis showed that retentostat-grown cells had significantly increased abundance of a variety of stress proteins involved in protein quality maintenance and DNA repair (DnaJ, Hsp90, FtsH, ClpB, ClpP1, ClpC, GroES, RuvB, RecA), as well as proteins involved in oxidative stress defence (peroxiredoxin, ferredoxin, thioredoxin peroxidase, glutaredoxin and thioredoxin reductase). Exposure to three different stress conditions, 45 °C, pH 3, and 10 mM H2O2, showed highest stress resistance of retentostat cells sampled at week 2 and week 3 grown at 0.0018 and 0.00081 h-1. Our findings show that cultivation at near-zero growth rates induces higher abundance of stress defence proteins contributing to the robustness of B. breve NRBB57, thereby offering an approach that may support its production and functionality.
Collapse
Affiliation(s)
- A R Ortiz Camargo
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - O Van Mastrigt
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - R S Bongers
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands
| | - K Ben-Amor
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands
| | - J Knol
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands.,Laboratory of Microbiology, Wageningen University & Research, P.O. Box 8033, 6700 EH Wageningen, the Netherlands
| | - E J Smid
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - T Abee
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
21
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
22
|
Derrien M, Mikulic N, Uyoga MA, Chenoll E, Climent E, Howard-Varona A, Nyilima S, Stoffel NU, Karanja S, Kottler R, Stahl B, Zimmermann MB, Bourdet-Sicard R. Gut microbiome function and composition in infants from rural Kenya and association with human milk oligosaccharides. Gut Microbes 2023; 15:2178793. [PMID: 36794816 PMCID: PMC9980514 DOI: 10.1080/19490976.2023.2178793] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
The gut microbiota evolves rapidly after birth, responding dynamically to environmental factors and playing a key role in short- and long-term health. Lifestyle and rurality have been shown to contribute to differences in the gut microbiome, including Bifidobacterium levels, between infants. We studied the composition, function and variability of the gut microbiomes of 6- to 11-month-old Kenyan infants (n = 105). Shotgun metagenomics showed Bifidobacterium longum to be the dominant species. A pangenomic analysis of B. longum in gut metagenomes revealed a high prevalence of B. longum subsp. infantis (B. infantis) in Kenyan infants (80%), and possible co-existence of this subspecies with B. longum subsp. longum. Stratification of the gut microbiome into community (GMC) types revealed differences in composition and functional features. GMC types with a higher prevalence of B. infantis and abundance of B. breve also had a lower pH and a lower abundance of genes encoding pathogenic features. An analysis of human milk oligosaccharides (HMOs) classified the human milk (HM) samples into four groups defined on the basis of secretor and Lewis polymorphisms revealed a higher prevalence of HM group III (Se+, Le-) (22%) than in most previously studied populations, with an enrichment in 2'-fucosyllactose. Our results show that the gut microbiome of partially breastfed Kenyan infants over the age of six months is enriched in bacteria from the Bifidobacterium community, including B. infantis, and that the high prevalence of a specific HM group may indicate a specific HMO-gut microbiome association. This study sheds light on gut microbiome variation in an understudied population with limited exposure to modern microbiome-altering factors.
Collapse
Affiliation(s)
- Muriel Derrien
- Advanced Health & Science, Danone Nutricia Research, Palaiseau, France,CONTACT Muriel Derrien Advanced Health & Science, Danone Nutricia Research, Palaiseau, France
| | - Nadja Mikulic
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Mary A Uyoga
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Empar Chenoll
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Eric Climent
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Adrian Howard-Varona
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Suzane Nyilima
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Simon Karanja
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | | | - Bernd Stahl
- Advanced Health & Science, Danone Nutricia Research, Utrecht, The Netherlands,Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Raphaëlle Bourdet-Sicard
- Advanced Health & Science, Danone Nutricia Research, Palaiseau, France,Raphaëlle Bourdet-Sicard Advanced Health & Science, Danone Nutricia Research, Palaiseau, France
| |
Collapse
|
23
|
Danilenko VN, Alekseeva MG, Koshenko TA, Kovtun AS, Nezametdinova VZ. Species-Forming PFNA Operon of Bifidobacteria: Modules of Sensor Proteins Pkb2 and FN3, Structure and Distribution among Different Species and Strains of Bifidobacteria in Human Intestinal Microbiome. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422090071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
24
|
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: 2.3] [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
|
25
|
Diamond L, Wine R, Morris SK. Impact of intrapartum antibiotics on the infant gastrointestinal microbiome: a narrative review. Arch Dis Child 2022; 107:627-634. [PMID: 34716171 DOI: 10.1136/archdischild-2021-322590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The composition of the infant gastrointestinal (GI) microbiome has been linked to adverse long-term health outcomes and neonatal sepsis. Several factors are known to impact the composition of the microbiome, including mode of delivery, gestational age, feeding method and exposure to antibiotics. The impact of intrapartum antibiotics (IPAs) on the infant microbiome requires further research. OBJECTIVE We aimed to evaluate the impact of IPAs on the infant GI microbiome. METHODS We searched Ovid MEDLINE and Embase Classic+Embase for articles in English reporting on the microbiome of infants exposed to IPAs from the date of inception to 3 January 2021. Primary outcomes included abundance and colonisation of Bifidobacterium and Lactobacillus, as well as alpha and beta diversity. RESULTS 30 papers were included in this review. In the first year of life, following exposure to IPAs, 30% (6/20) of infant cohorts displayed significantly reduced Bifidobacterium, 89% (17/19) did not display any significant differences in Lactobacillus colonisation, 21% (7/34) displayed significantly reduced alpha diversity and 35% (12/34) displayed alterations in beta diversity. Results were further stratified by delivery, gestational age (preterm or full term) and feeding method. CONCLUSIONS IPAs impact the composition of the infant GI microbiome, resulting in possible reductions Bifidobacterium and alpha diversity, and possible alterations in beta diversity. Our findings may have implications for maternal and neonatal health, including interventions to prevent reductions in health-promoting bacteria (eg, probiotics) and IPA class selection.
Collapse
Affiliation(s)
- Laura Diamond
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Wine
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaun K Morris
- Division of Infectious Diseases and Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada .,Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Gold MS, Quinn PJ, Campbell DE, Peake J, Smart J, Robinson M, O’Sullivan M, Vogt JK, Pedersen HK, Liu X, Pazirandeh-Micol E, Heine RG. Effects of an Amino Acid-Based Formula Supplemented with Two Human Milk Oligosaccharides on Growth, Tolerability, Safety, and Gut Microbiome in Infants with Cow's Milk Protein Allergy. Nutrients 2022; 14:nu14112297. [PMID: 35684099 PMCID: PMC9182596 DOI: 10.3390/nu14112297] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/04/2022] Open
Abstract
This open-label, non-randomized, multicenter trial (Registration: NCT03661736) aimed to assess if an amino acid-based formula (AAF) supplemented with two human milk oligosaccharides (HMO) supports normal growth and is well tolerated in infants with a cow's milk protein allergy (CMPA). Term infants aged 1-8 months with moderate-to-severe CMPA were enrolled. The study formula was an AAF supplemented with 2'-fucosyllactose (2'-FL) and lacto-N-neotetraose (LNnT). Infants were fed the study formula for 4 months and were offered to remain on the formula until 12 months of age. Tolerance and safety were assessed throughout the trial. Out of 32 infants (mean age 18.6 weeks; 20 (62.5%) male), 29 completed the trial. During the 4-month principal study period, the mean weight-for-age Z score (WAZ) increased from -0.31 at the baseline to +0.28 at the 4-months' follow-up. Linear and head growth also progressed along the WHO child growth reference, with a similar small upward trend. The formula was well tolerated and had an excellent safety profile. When comparing the microbiome at the baseline to the subsequent visits, there was a significant on-treatment enrichment in HMO-utilizing bifidobacteria, which was associated with a significant increase in fecal short-chain fatty acids. In addition, we observed a significant reduction in the abundance of fecal Proteobacteria, suggesting that the HMO-supplemented study formula partially corrected the gut microbial dysbiosis in infants with CMPA.
Collapse
Affiliation(s)
- Michael S. Gold
- Department of Allergy & Immunology, Women’s and Children’s Hospital, University of Adelaide, Adelaide, SA 5006, Australia;
- Correspondence:
| | - Patrick J. Quinn
- Department of Allergy & Immunology, Women’s and Children’s Hospital, University of Adelaide, Adelaide, SA 5006, Australia;
| | - Dianne E. Campbell
- Department of Allergy & Clinical Immunology, Children’s Hospital at Westmead, University of Sydney, Sydney, NSW 2145, Australia;
| | - Jane Peake
- Queensland Paediatric Immunology and Allergy Service, Queensland Children’s Hospital, University of Queensland, South Brisbane, QLD 4101, Australia;
| | - Joanne Smart
- Paediatric Allergy Services, Epworth Hospital, Richmond, VIC 3121, Australia;
| | - Marnie Robinson
- Melbourne Allergy Centre & Children’s Specialists Medical Group, Parkville, VIC 3152, Australia;
| | - Michael O’Sullivan
- Department of Immunology, Perth Children’s Hospital, Nedlands, WA 6009, Australia
| | | | | | - Xiaoqiu Liu
- Biostatistics and Data Science Division, The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia;
| | | | - Ralf G. Heine
- Nestlé Health Science, CH-1800 Vevey, Switzerland; (E.P.-M.); (R.G.H.)
| |
Collapse
|
27
|
Grant-Beurmann S, Jumare J, Ndembi N, Matthew O, Shutt A, Omoigberale A, Martin OA, Fraser CM, Charurat M. Dynamics of the infant gut microbiota in the first 18 months of life: the impact of maternal HIV infection and breastfeeding. MICROBIOME 2022; 10:61. [PMID: 35414043 PMCID: PMC9004197 DOI: 10.1186/s40168-022-01230-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Access to antiretroviral therapy (ART) during pregnancy and breastfeeding for mothers with HIV has resulted in fewer children acquiring HIV peri- and postnatally, resulting in an increase in the number of children who are exposed to the virus but are not infected (HEU). HEU infants have an increased likelihood of childhood infections and adverse growth outcomes, as well as increased mortality compared to their HIV-unexposed (HUU) peers. We explored potential differences in the gut microbiota in a cohort of 272 Nigerian infants born to HIV-positive and negative mothers in this study during the first 18 months of life. RESULTS The taxonomic composition of the maternal vaginal and gut microbiota showed no significant differences based on HIV status, and the composition of the infant gut microbiota at birth was similar between HUU and HEU. Longitudinal taxonomic composition of the infant gut microbiota and weight-for-age z-scores (WAZ) differed depending on access to breast milk. HEU infants displayed overall lower WAZ than HUU infants at all time points. We observed a significantly lower relative abundance of Bifidobacterium in HEU infants at 6 months postpartum. Breast milk composition also differed by time point and HIV infection status. The antiretroviral therapy drugs, lamivudine and nevirapine, as well as kynurenine, were significantly more abundant in the breast milk of mothers with HIV. Levels of tiglyl carnitine (C5) were significantly lower in the breast milk of mothers without HIV. ART drugs in the breast milk of mothers with HIV were associated with a lower relative abundance of Bifidobacterium longum. CONCLUSIONS Maternal HIV infection was associated with adverse growth outcomes of HEU infants in this study, and these differences persist from birth through at least 18 months, which is a critical window for the development of the immune and central nervous systems. We observed that the relative abundance of Bifidobacterium spp. was significantly lower in the gut microbiota of all HEU infants over the first 6 months postpartum, even if HEU infants were receiving breast milk. Breastfeeding was of benefit in our HEU infant cohort in the first weeks postpartum; however, ART drug metabolites in breast milk were associated with a lower abundance of Bifidobacterium. Video abstract.
Collapse
Affiliation(s)
- Silvia Grant-Beurmann
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jibreel Jumare
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Ashley Shutt
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Olivia A Martin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Man Charurat
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Sharma S, Singh S, Chaudhary V, Mantri S, Chander A, Maurya R, Rajarammohan S, Singh RP, Rishi P, Bishnoi M, Bhadada SK, Kondepudi KK. Isomaltooligosaccharides utilization and genomic characterization of human infant anti-inflammatory Bifidobacterium longum and Bifidobacterium breve strains. 3 Biotech 2022; 12:89. [PMID: 35299989 PMCID: PMC8901852 DOI: 10.1007/s13205-022-03141-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/07/2022] [Indexed: 11/01/2022] Open
Abstract
This study was carried out to understand the probiotic features, ability to utilize non-digestible carbohydrates and comparative genomics of anti-inflammatory Bifidobacterium strains isolated from human infant stool samples. Bacterial strains were isolated from the stool samples using serial dilution on MRS agar plates supplemented with 0.05% l-cysteine hydrochloride and mupirocin. Molecular characterization of the strains was carried out by 16S rRNA gene sequencing. Anti-inflammatory activity was determined using TNF-α and lipopolysaccharide (LPS) induced inflammation in Caco2 cells. Probiotic attributes were determined as per the established protocols. Isomaltooligosaccharides (IMOS) utilization was determined in the broth cultures. Whole genome sequencing and analysis was carried out for three strains. Four obligate anaerobic, Gram positive Bifidobacterium strains were isolated from the infant stool samples. Strains were identified as Bifidobacterium longum Bif10, B. breve Bif11, B. longum Bif12 and B. longum Bif16. The strains were able to prevent inflammation in the Caco2 cells through lowering of IL8 production that was caused by TNF-α and LPS treatment. The strains exhibited desirable probiotic attributes such as acid and bile tolerance, mucin binding, antimicrobial activity, bile salt hydrolase activity, cholesterol lowering ability and could ferment non-digestible carbohydrates such as isomaltooligosaccharides and raffinose. Furthermore, Isomaltooligosaccharides supported the optimum growth of the strains in vitro, which was comparable to that on glucose. Strains could metabolize IMOS through cell associated α-glucosidase activity. Genomic features revealed the presence of genes responsible for the utilization of IMOS and for the probiotic attributes. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03141-2.
Collapse
|
29
|
Zhao X, Hogenkamp A, Li X, Chen H, Garssen J, Knippels LMJ. Role of selenium in IgE mediated soybean allergy development. Crit Rev Food Sci Nutr 2022; 63:7016-7024. [PMID: 35187987 DOI: 10.1080/10408398.2022.2039898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Food allergy is a pathological immune reaction triggered by normal innocuous dietary proteins. Soybean is widely used in many food products and has long been recognized as a source of high-quality proteins. However, soybean is listed as one of the 8 most significant food allergens. The prevalence of soybean allergy is increasing worldwide and impacts the quality of life of patients. Currently, the only strategy to manage food allergy relies on strict avoidance of the offending food. Nutritional supplementation is a new prevention strategy which is currently under evaluation. Selenium (Se), as one of the essential micronutrients for humans and animals, carries out biological effects through its incorporation into selenoproteins. The use of interventions with micronutrients, like Se, might be an interesting new approach. In this review we describe the involvement of Se in a variety of processes, including maintaining immune homeostasis, preventing free radical damage, and modulating the gut microbiome, all of which may contribute to in both the prevention and treatment of food allergy. Se interventions could be an interesting new approach for future treatment strategies to manage soybean allergy, and food allergy in general, and could help to improve the quality of life for food allergic patients.
Collapse
Affiliation(s)
- Xiaoli Zhao
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- State Key Laboratory of Food Science and Technology, Nanchang University, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Global Centre of Excellence Immunology, Danone/Nutricia Research, Utrecht, The Netherlands
| | - Leon M J Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Global Centre of Excellence Immunology, Danone/Nutricia Research, Utrecht, The Netherlands
| |
Collapse
|
30
|
Vandenplas Y, Żołnowska M, Berni Canani R, Ludman S, Tengelyi Z, Moreno-Álvarez A, Goh AEN, Gosoniu ML, Kirwan BA, Tadi M, Heine RG. Effects of an Extensively Hydrolyzed Formula Supplemented with Two Human Milk Oligosaccharides on Growth, Tolerability, Safety and Infection Risk in Infants with Cow's Milk Protein Allergy: A Randomized, Multi-Center Trial. Nutrients 2022; 14:530. [PMID: 35276889 PMCID: PMC8839689 DOI: 10.3390/nu14030530] [Citation(s) in RCA: 15] [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: 12/22/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
This randomized clinical trial (Registration: NCT03085134) assessed if an extensively hydrolyzed formula (EHF) supplemented with two human milk oligosaccharides (HMO) and reduced protein content (2.20 g/100 kcal) supports normal growth in infants with cow’s milk protein allergy (CMPA). Secondary outcomes were gastrointestinal tolerability, safety, and effect on infections. Nonbreastfed infants aged 0−6 months with CMPA were enrolled. Body weight, length, and head circumference were measured monthly for 4 months (primary study endpoint), after 6 months, and at the age of 12 months. Of 200 infants screened, 194 (mean age 3.2 months) were randomized. At the 4-month follow-up, daily weight gain for the test formula was noninferior to the control formula; p < 0.005. There were no significant group differences in anthropometric parameters. Both formulas were safe and well tolerated. Infants in the HMO group had a statistically significant reduction in the frequency of upper respiratory tract infections and a lower incidence of ear infections at 12 months (per protocol analysis). The relative risk of lower respiratory tract and gastrointestinal infections was reduced by 30−40%, but this was not statistically significant due to sample size limitations. In summary, the HMO-supplemented formula supports normal growth in infants with CMPA and suggests a protective effect against respiratory and ear infections in the first year of life.
Collapse
Affiliation(s)
- Yvan Vandenplas
- UZ Brussel KidZ Health Castle, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | - Roberto Berni Canani
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy;
| | - Siân Ludman
- Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK;
| | | | - Ana Moreno-Álvarez
- Department of Pediatrics, A Coruña University Hospital, 15006 A Coruña, Spain;
| | - Anne E. N. Goh
- KK Women’s and Children’s Hospital, Singapore 229899, Singapore;
| | | | | | - Monika Tadi
- Nestlé Health Science, 1800 Vevey, Switzerland; (M.T.); (R.G.H.)
| | - Ralf G. Heine
- Nestlé Health Science, 1800 Vevey, Switzerland; (M.T.); (R.G.H.)
| | | |
Collapse
|
31
|
Sprenger N, Tytgat HL, Binia A, Austin S, Singhal A. Biology of human milk oligosaccharides: from Basic Science to Clinical Evidence. J Hum Nutr Diet 2022; 35:280-299. [PMID: 35040200 PMCID: PMC9304252 DOI: 10.1111/jhn.12990] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Human milk oligosaccharides (HMOs) have been researched by scientists for over 100 years, driven by the substantial evidence for the nutritional and health benefits of mother's milk. Yet research has truly bloomed during the last decade, thanks to the progress in biotechnology, which allowed the production of large amounts of bona fide HMOs. The availability of HMOs has been particularly crucial for the renewed interest in HMO research because of the low abundance or even absence of HMOs in farmed animal milk. This interest is reflected in the increasing number of original research publications and reviews on HMOs. Here, we provide an overview and critical discussion on structure function relations of HMOs that highlight why they are such interesting and important components of human milk. Clinical observations in breastfed infants backed by basic research from animal models provide guidance as to what physiological roles for HMOs are to be expected. From an evidence-based nutrition viewpoint, we discuss the current data supporting clinical relevance of specific HMOs based on randomized placebo controlled clinical intervention trials in formula-fed infants. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Hanne Lp Tytgat
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Aristea Binia
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sean Austin
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Atul Singhal
- Institute of Child Health, University College London, London, WC1N 1EH, United Kingdom
| |
Collapse
|
32
|
The Effectiveness and Safety of Probiotic Supplements for Psoriasis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials and Preclinical Trials. J Immunol Res 2021; 2021:7552546. [PMID: 34938815 PMCID: PMC8687811 DOI: 10.1155/2021/7552546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/05/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Background Patients with psoriasis need long-term medication to control their condition. Recent studies suggest that changing the intestinal flora may be a potential treatment. Methods The databases were utilized to search the randomized controlled trials (RCTs) and preclinical trials about probiotic supplement in the treatment of psoriasis. The retrieval time is from the establishment of these databases to December 2020. RevMan5.3 was used for the risk assessment of bias and meta-analysis. This systematic review was registered in PROSPERO (CRD42021232756). Results A total of 3 RCTs involving 164 participants were included. Two RCTs showed that probiotics can improve PASI and thereby improve the condition. For inflammation-related indicators, only one RCT showed that probiotics can improve the levels of CRP and TNF-α but have no obvious improvement effect on IL6. One RCT demonstrated the total effective rate of probiotics in the treatment of psoriasis. For adverse events, one RCT showed that the incidence of adverse events of probiotic treatment was low. Preclinical studies showed that continuous intervention with oral probiotics can significantly improve the progression of psoriasis and reduce the expression of inflammatory factors. The meta-analysis showed that the PASI between two groups was of no statistical significance (SMD 1.83 [-0.41, 4.07], P = 0.11). Meanwhile, probiotics may improve skin thickness (SMD -5.87 [-11.34, -0.41], P = 0.04) in animal model. Conclusion Prebiotics may have a positive effect on alleviating the clinical symptoms of psoriasis, but a large sample of RCTs is still needed to support its therapeutic effect in psoriasis.
Collapse
|
33
|
Saturio S, Nogacka AM, Alvarado-Jasso GM, Salazar N, de los Reyes-Gavilán CG, Gueimonde M, Arboleya S. Role of Bifidobacteria on Infant Health. Microorganisms 2021; 9:2415. [PMID: 34946017 PMCID: PMC8708449 DOI: 10.3390/microorganisms9122415] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/19/2022] Open
Abstract
Bifidobacteria are among the predominant microorganisms during infancy, being a dominant microbial group in the healthy breastfed infant and playing a crucial role in newborns and infant development. Not only the levels of the Bifidobacterium genus but also the profile and quantity of the different bifidobacterial species have been demonstrated to be of relevance to infant health. Although no definitive proof is available on the causal association, reduced levels of bifidobacteria are perhaps the most frequently observed alteration of the intestinal microbiota in infant diseases. Moreover, Bifidobacterium strains have been extensively studied by their probiotic attributes. This review compiles the available information about bifidobacterial composition and function since the beginning of life, describing different perinatal factors affecting them, and their implications on different health alterations in infancy. In addition, this review gathers exhaustive information about pre-clinical and clinical studies with Bifidobacterium strains as probiotics in neonates.
Collapse
Affiliation(s)
- Silvia Saturio
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Guadalupe M. Alvarado-Jasso
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
34
|
Effect of Intrapartum Antibiotics Prophylaxis on the Bifidobacterial Establishment within the Neonatal Gut. Microorganisms 2021; 9:microorganisms9091867. [PMID: 34576761 PMCID: PMC8471514 DOI: 10.3390/microorganisms9091867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022] Open
Abstract
Antibiotics are important disruptors of the intestinal microbiota establishment, linked to immune and metabolic alterations. The intrapartum antibiotics prophylaxis (IAP) is a common clinical practice that is present in more than 30% of labours, and is known to negatively affect the gut microbiota composition. However, little is known about how it affects to Bifidobacterium (sub)species level, which is one of the most important intestinal microbial genera early in life. This study presents qualitative and quantitative analyses of the bifidobacterial (sub)species populations in faecal samples, collected at 2, 10, 30 and 90 days of life, from 43 healthy full-term babies, sixteen of them delivered after IAP use. This study uses both 16S rRNA–23S rRNA internal transcribed spacer (ITS) region sequencing and q-PCR techniques for the analyses of the relative proportions and absolute levels, respectively, of the bifidobacterial populations. Our results show that the bifidobacterial populations establishment is affected by the IAP at both quantitative and qualitative levels. This practice can promote higher bifidobacterial diversity and several changes at a compositional level. This study underlines specific targets for developing gut microbiota-based products for favouring a proper bifidobacterial microbiota development when IAP is required.
Collapse
|
35
|
Nezametdinova VZ, Yunes RA, Dukhinova MS, Alekseeva MG, Danilenko VN. The Role of the PFNA Operon of Bifidobacteria in the Recognition of Host's Immune Signals: Prospects for the Use of the FN3 Protein in the Treatment of COVID-19. Int J Mol Sci 2021; 22:ijms22179219. [PMID: 34502130 PMCID: PMC8430577 DOI: 10.3390/ijms22179219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Bifidobacteria are some of the major agents that shaped the immune system of many members of the animal kingdom during their evolution. Over recent years, the question of concrete mechanisms underlying the immunomodulatory properties of bifidobacteria has been addressed in both animal and human studies. A possible candidate for this role has been discovered recently. The PFNA cluster, consisting of five core genes, pkb2, fn3, aaa-atp, duf58, tgm, has been found in all gut-dwelling autochthonous bifidobacterial species of humans. The sensory region of the species-specific serine-threonine protein kinase (PKB2), the transmembrane region of the microbial transglutaminase (TGM), and the type-III fibronectin domain-containing protein (FN3) encoded by the I gene imply that the PFNA cluster might be implicated in the interaction between bacteria and the host immune system. Moreover, the FN3 protein encoded by one of the genes making up the PFNA cluster, contains domains and motifs of cytokine receptors capable of selectively binding TNF-α. The PFNA cluster could play an important role for sensing signals of the immune system. Among the practical implications of this finding is the creation of anti-inflammatory drugs aimed at alleviating cytokine storms, one of the dire consequences resulting from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Venera Z. Nezametdinova
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Roman A. Yunes
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Marina S. Dukhinova
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, 197101 Saint-Petersburg, Russia;
| | - Maria G. Alekseeva
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Valery N. Danilenko
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
- Correspondence:
| |
Collapse
|
36
|
Greaves M, Cazzaniga V, Ford A. Can we prevent childhood Leukaemia? Leukemia 2021; 35:1258-1264. [PMID: 33833382 PMCID: PMC8102184 DOI: 10.1038/s41375-021-01211-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/03/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Valeria Cazzaniga
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Anthony Ford
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| |
Collapse
|
37
|
Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. Breast Milk: A Source of Functional Compounds with Potential Application in Nutrition and Therapy. Nutrients 2021; 13:1026. [PMID: 33810073 PMCID: PMC8005182 DOI: 10.3390/nu13031026] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is an unbeatable food that covers all the nutritional requirements of an infant in its different stages of growth up to six months after birth. In addition, breastfeeding benefits both maternal and child health. Increasing knowledge has been acquired regarding the composition of breast milk. Epidemiological studies and epigenetics allow us to understand the possible lifelong effects of breastfeeding. In this review we have compiled some of the components with clear functional activity that are present in human milk and the processes through which they promote infant development and maturation as well as modulate immunity. Milk fat globule membrane, proteins, oligosaccharides, growth factors, milk exosomes, or microorganisms are functional components to use in infant formulas, any other food products, nutritional supplements, nutraceuticals, or even for the development of new clinical therapies. The clinical evaluation of these compounds and their commercial exploitation are limited by the difficulty of isolating and producing them on an adequate scale. In this work we focus on the compounds produced using milk components from other species such as bovine, transgenic cattle capable of expressing components of human breast milk or microbial culture engineering.
Collapse
Affiliation(s)
- Cristina Sánchez
- Pharmacy Faculty, San Pablo-CEU University, 28003 Madrid, Spain;
| | - Luis Franco
- Medicine Faculty, Santiago de Compostela University, 15782 Santiago de Compostela, Spain;
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| |
Collapse
|
38
|
Alessandri G, van Sinderen D, Ventura M. The genus bifidobacterium: From genomics to functionality of an important component of the mammalian gut microbiota running title: Bifidobacterial adaptation to and interaction with the host. Comput Struct Biotechnol J 2021; 19:1472-1487. [PMID: 33777340 PMCID: PMC7979991 DOI: 10.1016/j.csbj.2021.03.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Members of the genus Bifidobacterium are dominant and symbiotic inhabitants of the mammalian gastrointestinal tract. Being vertically transmitted, bifidobacterial host colonization commences immediately after birth and leads to a phase of host infancy during which bifidobacteria are highly prevalent and abundant to then transit to a reduced, yet stable abundance phase during host adulthood. However, in order to reach and stably colonize their elective niche, i.e. the large intestine, bifidobacteria have to cope with a multitude of oxidative, osmotic and bile salt/acid stress challenges that occur along the gastrointestinal tract (GIT). Concurrently, bifidobacteria not only have to compete with the myriad of other gut commensals for nutrient acquisition, but they also require protection against bacterial viruses. In this context, Next-Generation Sequencing (NGS) techniques, allowing large-scale comparative and functional genome analyses have helped to identify the genetic strategies that bifidobacteria have developed in order to colonize, survive and adopt to the highly competitive mammalian gastrointestinal environment. The current review is aimed at providing a comprehensive overview concerning the molecular strategies on which bifidobacteria rely to stably and successfully colonize the mammalian gut.
Collapse
Affiliation(s)
- Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Ireland and School of Microbiology, University College Cork, Western Road, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
39
|
Phavichitr N, Wang S, Chomto S, Tantibhaedhyangkul R, Kakourou A, Intarakhao S, Jongpiputvanich S, Roeselers G, Knol J. Impact of synbiotics on gut microbiota during early life: a randomized, double-blind study. Sci Rep 2021; 11:3534. [PMID: 33574421 PMCID: PMC7878856 DOI: 10.1038/s41598-021-83009-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
Human milk is considered the optimal nutrition for infants and found to contain significant numbers of viable bacteria. The aim of the study was to assess the effects of a specific synbiotic combination at doses closer to the bacterial cells present in human milk, on intestinal bifidobacteria proportions (relative abundance), reduction of potential pathogens and gut physiological conditions. A clinical study was conducted in 290 healthy infants aged from 6 to 19 weeks. Infants received either a control infant formula or one of the two investigational infant formulas (control formula with 0.8 g/100 ml scGOS/lcFOS and Bifidobacterium breve M-16V at either 1 × 104 cfu/ml or 1 × 106 cfu/ml). Exclusively breastfed infants were included as a reference. Analyses were performed on intention-to-treat groups and all-subjects-treated groups. After 6 weeks of intervention, the synbiotics at two different doses significantly increased the bifidobacteria proportions in healthy infants. The synbiotic supplementation also decreased the prevalence (infants with detectable levels) and the abundance of C. difficile. Closer to the levels in the breastfed reference group, fecal pH was significantly lower while l-lactate concentrations and acetate proportions were significantly higher in the synbiotic groups. All formulas were well tolerated and all groups showed a comparable safety profile based on the number and severity of adverse events and growth. In healthy infants, supplementation of infant-type bifidobacterial strain B. breve M-16V, at a dose close to bacterial numbers found in human milk, with scGOS/lcFOS (9:1) created a gut environment closer to the breastfed reference group. This specific synbiotic mixture may also support gut microbiota resilience during early life. Clinical Trial Registration This clinical study named Color Synbiotics Study, was registered in ClinicalTrials.gov on 18 March 2013. Registration number is NCT01813175. https://clinicaltrials.gov/ct2/show/NCT01813175.
Collapse
Affiliation(s)
| | - Shugui Wang
- Danone Nutricia Research, Singapore, Singapore
| | - Sirinuch Chomto
- Nutritional Unit, Department of Pediatrics, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Sukkrawan Intarakhao
- Department of Pediatrics, Thammasat Hospital, Faculty of Medicine, Thammasat University, Bangkok, Thailand
| | - Sungkom Jongpiputvanich
- Department of Pediatrics, Thammasat Hospital, Faculty of Medicine, Thammasat University, Bangkok, Thailand
| | | | | | - Jan Knol
- Danone Nutricia Research, Utrecht, The Netherlands.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|