1
|
Bauer Estrada K, Conde-Martínez N, Acosta-González A, Díaz-Barrera LE, Rodríguez-Castaño GP, Quintanilla-Carvajal MX. Synbiotics of encapsulated Limosilactobacillus fermentum K73 promotes in vitro favorable gut microbiota shifts and enhances short-chain fatty acid production in fecal samples of children with autism spectrum disorder. Food Res Int 2025; 209:116227. [PMID: 40253179 DOI: 10.1016/j.foodres.2025.116227] [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: 11/21/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Modulation of the gut microbiota has emerged as a promising approach for addressing the gastrointestinal and neurodevelopmental symptoms associated with autism spectrum disorder (ASD). Consequently, this study aimed to evaluate the impact of four formulated synbiotics comprising Limoscilactobacillus fermentum K73, high-oleic palm oil and whey, on the gut microbiota composition of Colombian children with and without ASD. These components were encapsulated through high-shear emulsification and spray drying. The four synbiotics and their individual components were subjected to in vitro digestion and fermentation using samples of Colombian children gut microbiota. Short-chain fatty acids (SCFAs), including lactic, acetic, propionic, and butyric acids, were quantified using HPLC-DAD, while serotonin was determined by an ELISA kit after in vitro fermentations. Changes in microbial structure were assessed by the sequencing of the 16S rRNA gene via next-generation sequencing (NGS). The results revealed a decrease in the abundance of genera like Bacteroides and Dorea in ASD-associated samples after the treatment with the synbiotics. Conversely, an increase in the relative abundance of probiotic-related genera, including Lactobacillus, Streptococcus, and Anaerostipes, was observed. Furthermore, the analysis of SCFAs and serotonin indicated that the synbiotic intervention resulted in an elevated butyric acid and microbial serotonin synthesis, alongside a decrease in propionic acid, which is changes considered beneficial in the context of ASD. This evidence suggests that synbiotics of L. fermentum K73 could represent a promising live biotherapeutic strategy for modulating the gut microbiota of children with ASD.
Collapse
Affiliation(s)
- Katherine Bauer Estrada
- Biosciences Doctorate, Engineering Department, Universidad de La Sabana, Chía, Colombia; Engineering Department, Universidad de La Sabana, Chía, Colombia
| | | | - Alejandro Acosta-González
- Engineering Department, Universidad de La Sabana, Chía, Colombia; Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chía, Colombia
| | | | | | - María Ximena Quintanilla-Carvajal
- Engineering Department, Universidad de La Sabana, Chía, Colombia; Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chía, Colombia.
| |
Collapse
|
2
|
Gianotti A, Marin V, Cardone G, Bordoni A, Mancini E, Magni M, Pichler A, Ciani S, Polenghi O, Cerne VL, Nissen L. Personalized and precise functional assessment of innovative flatbreads toward the colon microbiota of people with metabolic syndrome: Results from an in vitro simulation. Food Res Int 2025; 209:116197. [PMID: 40253173 DOI: 10.1016/j.foodres.2025.116197] [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: 10/30/2024] [Revised: 02/20/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Due to the increasing incidence of individuals with metabolic syndrome and the higher correlations between metabolic syndrome and the gut microbiota, there is a need to formulate functional foods that can promote the development of beneficial microorganisms within the gut microbiota. This study aims to evaluate the possible positive effects of innovative gluten-free flatbread, containing grape and apple antioxidant-rich by-products, on the gut microbiota of patients with metabolic syndrome. The baked products were subjected to gastric digestion using the Infogest system, followed by proximal colonic fermentation in the MICODE (Multi-Unit In vitro Colon Model) intestinal model, where three samplings were performed (baseline, after 16 h and 24 h). The samples were then subjected to 16S rRNA metataxonomy, quantification of shifts in bacterial populations by qPCR analysis and characterization of volatile organic compounds by SPME GC-MS (Solid Phase Micro Extraction Gas-Chromatography Mass-Spectrophotometry). A robust statistical approach based on several tests and multivariate analysis was applied. The results obtained demonstrated the in vitro potential of functional flatbreads in improving the dysbiosis of the microbiota of individuals with metabolic syndrome, due to a reduction in the Firmicutes/Bacteroidota ratio, and highlighted an increase in commensal microorganisms (Bifidobacterium, positive clostridia and Akkermansia muciniphila) and a reduction in negative microorganisms (Enterobacteriaceae, negative clostridia and Collinsella spp.). The analysis of metabolites showed an increase in health-beneficial metabolites (acetate and medium chain organic acids) and a reduction in harmful metabolites (p-cresol and skatole), the degree of this modulation varied based on the flatbread composition. While this study employed an in vitro model of recognized limitations, it nonetheless provides valuable, evidence-based results that can be used for preclinical screening of formulations. Anyhow this work is of high fashion in this running time as it proposes i) in vitro models rather than animal testing; ii) human MetS gut microbiota for high translatability to in vivo condition; iii) approaches of precise and personalized nutrition by the use of specific microbiota and omic technologies, all component that vow to be the future of food assessment.
Collapse
Affiliation(s)
- Andrea Gianotti
- DiSTAL - Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Food Science Campus, P.za G. Goidanich, 60, 47521 Cesena, Italy; CIRI - Interdepartmental Centre of Agri-Food Industrial Research, Alma Mater Studiorum - University of Bologna, P.za G. Goidanich, 60, 47521 Cesena, Italy..
| | - Veronica Marin
- Dr. Schär R&D Centre, Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Gaetano Cardone
- Dr. Schär R&D Centre, Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Alessandra Bordoni
- DiSTAL - Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Food Science Campus, P.za G. Goidanich, 60, 47521 Cesena, Italy; CIRI - Interdepartmental Centre of Agri-Food Industrial Research, Alma Mater Studiorum - University of Bologna, P.za G. Goidanich, 60, 47521 Cesena, Italy
| | - Elisa Mancini
- Private Nutritional Clinic, Piazza Polesani nel Mondo, 16, 45100, Boara Polesine (RO), Italy
| | | | | | - Silvano Ciani
- Dr. Schär R&D Centre, Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Ombretta Polenghi
- Dr. Schär R&D Centre, Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Virna Lucia Cerne
- Dr. Schär R&D Centre, Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Lorenzo Nissen
- DiSTAL - Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Food Science Campus, P.za G. Goidanich, 60, 47521 Cesena, Italy; CIRI - Interdepartmental Centre of Agri-Food Industrial Research, Alma Mater Studiorum - University of Bologna, P.za G. Goidanich, 60, 47521 Cesena, Italy
| |
Collapse
|
3
|
Wang Y, Hu Q, Chen B, Ma D. Effects of Liupao Tea with Different Years of Aging on Glycolipid Metabolism, Body Composition, and Gut Microbiota in Adults with Obesity or Overweight: A Randomized, Double-Blind Study. Foods 2025; 14:866. [PMID: 40077569 PMCID: PMC11898661 DOI: 10.3390/foods14050866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Liupao tea (LPT) is a traditionally fermented dark tea from Guangxi, China and the effects of different aging periods of LPT on metabolic health remain inadequately explored. METHODS This randomized, double-blind, longitudinal study enrolled 106 adults with obesity or overweight who were assigned to consume LPT of different ages over a 90-day period. Participants were randomly divided into four groups, each consuming LPT that had been aged for 1 year, 4 years, 7 years, or 10 years. The metabolic parameters, body composition, and gut microbiota were assessed at baseline and after the 90-day intervention. RESULTS All LPT groups experienced significant reductions in systolic blood pressure (SBP) and diastolic blood pressure (DBP), with the 10-year-aged group showing the most notable SBP decrease (p < 0.001). Total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels decreased significantly in the 1-, 4-, and 10-year-aged groups (p < 0.05), while high-density lipoprotein cholesterol (HDL-C) increased in the 7-year-aged group (p < 0.05). Body weight, body fat mass (BFM), body mass index (BMI), waist circumference (WC), body fat percentage (BFP), and visceral fat area (VFA) significantly declined across all groups (p < 0.05). Gut microbiota analysis showed changes in specific genera, though overall diversity remained stable. No significant differences were found in metabolic or microbiota outcomes between the different aged groups. CONCLUSIONS LPT consumption effectively improves blood pressure, lipid profiles, and body composition in adults with obesity without adverse liver effects. The aging duration of LPT does not significantly alter these health benefits, challenging the belief that longer-aged LPT is superior.
Collapse
Affiliation(s)
| | | | | | - Defu Ma
- Department of Social Medicine and Health Education, School of Public Health, Peking University Health Science Center, Beijing 100191, China (Q.H.)
| |
Collapse
|
4
|
Taşkoparan Ş, Altınay C, Barbaros Özer H. Recent updates of probiotic dairy-based beverages. Food Funct 2025; 16:1656-1669. [PMID: 39962909 DOI: 10.1039/d4fo06322h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
There is a rapid paradigm shift in the food consumption habits of consumers globally. The interest in healthier, safer, minimally processed and nature-identical foods is the driving force of this paradigm shift. Although the roots of this consumer trend go back further, especially the Covid-19 pandemic has contributed to the acceleration of this process. The effects of probiotics on human health have been known for many years. The commercial success of some probiotic microorganism strains, supported by clinical studies, is also evident. Probiotic microorganisms can be found in commercial products in a wide range of forms including powder, tablets or incorporated into liquid or solid food matrices. Milk and dairy products are suitable vehicles for the delivery of probiotics into the human body. Apart from well-established dairy-based probiotic foods including yogurt and yogurt-type beverages, in recent years some dairy products supplemented or enhanced with postbiotics and paraprobiotics are gaining popularity. The incorporation of next-generation probiotics in probiotic beverage formulations has also attracted the attention of researchers. The current state-of-the art for the utilization of next-generation probiotics, postbiotics and paraprobiotics in dairy-based probiotic beverages is the main focus of this review. Conventional milk-, whey- and buttermilk-based probiotic beverages are also covered.
Collapse
Affiliation(s)
- Şevval Taşkoparan
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| | - Canan Altınay
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| | - H Barbaros Özer
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| |
Collapse
|
5
|
Wang W, Liu X, Nan H, Li H, Yan L. Specific gut microbiota and serum metabolite changes in patients with osteoarthritis. Front Cell Dev Biol 2025; 13:1543510. [PMID: 40027098 PMCID: PMC11868077 DOI: 10.3389/fcell.2025.1543510] [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: 12/11/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Recent research indicated a strong link between the gut microbiota and osteoarthritis. However, the complex interplay between the gut microbiota, serum metabolites, and the progression of osteoarthritis in affected individuals remains largely unexplored. This study aimed to investigate the characteristics of the gut microbiota and serum metabolites in patients with osteoarthritis. Methods Participants with either healthy knees or osteoarthritis were enrolled and categorized into healthy control (HC) and osteoarthritis (OA) groups. Fecal and blood samples were collected for 16S rRNA gene sequencing, metabolomic analysis via liquid chromatography-mass spectrometry (LC-MS), and integrated evaluation. Results The results showed no significant variation in gut microbiota richness and diversity between the two groups. However, the abundance of Bacteroides plebeius and Faecalibacterium prausnitzii was reduced in the OA group, both of which are known for their potential as next-generation probiotics for human health. Metabolomic analysis indicated that serum metabolites, including pyrogallol and 3-hydroxybutyrate (3HB), were significantly lower in the OA group. These metabolites are known to positively impact osteoarthritis progression and other diseases and demonstrated good diagnostic performance for distinguishing osteoarthritis patients from healthy controls. Correlation analysis revealed a positive correlation between Bacteroides plebeius and Faecalibacterium prausnitzii and between pyrogallol and 3HB. Discussion This study highlighted specific gut microbiota and serum metabolite profiles in osteoarthritis patients, suggesting that the specific changes in bacteria and derived metabolites are closely tied to osteoarthritis progression. This underscores the potential of gut microbiota and serum metabolites as modifiable elements and therapeutic targets for osteoarthritis prevention.
Collapse
Affiliation(s)
- Wendong Wang
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Xincheng Liu
- Department of Articular Orthopaedics, The Second People’s Hospital of Dalian, Dalian, China
| | - Hao Nan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Huan Li
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Litao Yan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
6
|
Argentino ICV, Godoy MG, Seldin L, Jurelevicius D. Distribution of Bacillota in Water and Sediments from Aquatic Environments. MICROBIAL ECOLOGY 2025; 88:3. [PMID: 39937305 PMCID: PMC11821768 DOI: 10.1007/s00248-024-02482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025]
Abstract
The phylum Bacillota comprises metabolically diverse bacteria with potential relevance in several fields. Although some genera, such as Bacillus and others, have been extensively studied, the distribution of Bacillota in the environment is still poorly understood. This study aimed to analyze the distribution of Bacillota present in water and sediment samples from different environments. For this purpose, water (W) and sediment (S) samples were collected from different marine environments (Dois Rios Beach - DR, Abraão Beach - AB, Massambaba Beach - MB, and Guanabara Bay - GB), freshwater lagoons (Jacarepiá Lagoon- JL) and hypersaline lagoons (Vermelha Lagoon - VL), all of which are located in Rio de Janeiro, Brazil. The microbial communities present in each sample were determined by sequencing 16S rRNA-encoding genes. The distribution and diversity of Bacillota were analyzed via QIIME2. The results revealed that Bacillota represented an average of 1% of the microbial community of aquatic microbiomes and were unevenly distributed in aquatic water and sediment. The highest abundances of Bacillota were detected in JL_S and VL_S, and the lowest abundances were observed in MB_W and AB_W. Only sequences related to the Bacilli and Clostridia classes were identified. The main Bacillota genera identified were Bacillus and an unidentified Clostridiales order genus. The VL_S and JL_S samples had the highest numbers of exclusive Bacillota genera. On the other hand, 15 Bacillota genera, which are generally observed in the human and animal guts, were found only in anthropogenically impacted GB_W and AB_W. The obtained results revealed how Bacillota are distributed in different aquatic environments.
Collapse
Affiliation(s)
- Isabella C V Argentino
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil
| | - Mateus G Godoy
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil
| | - Lucy Seldin
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil
| | - Diogo Jurelevicius
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
Xavier-Santos D, Bedani R, de Almeida Vieira I, Padilha M, Lima CMG, Silva JDR, Ferreira BM, Giraldo PC, Pagnossa JP, Sivieri K, Antunes AEC, Sant'Ana AS. Exploring the Potential Use of Probiotics, Prebiotics, Synbiotics, and Postbiotics as Adjuvants for Modulating the Vaginal Microbiome: a Bibliometric Review. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10444-8. [PMID: 39821884 DOI: 10.1007/s12602-024-10444-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Women's health is related to several factors that include physical, mental, and reproductive health. Additionally, the vaginal microbiota modulation performs a fundamental role in the regulation of physiological homeostasis and dysbiosis, which provides us a potential overview of the use of different biotic agents and their implications for female health. The objective of this work was propitiated insights and conception about the influence of probiotics, prebiotics, synbiotics, and postbiotics as adjuvants for prevention/treatment on the main infections that can affect women's health. Therefore, seventy-one studies published in the Web of Science Core Collection database from 1999 to 2024 were evaluated and performed to a bibliometric analysis employing the VOSviewer software for scientific mapping and network analysis. Our results suggest that administration of biotic agents as adjuvants are relevant for the prevention and/or treatment of the main diseases that affect female health, since they contribute to a healthy vaginal microbiota through anti-inflammatory and antimicrobial activities. Most clinical studies have demonstrated the effectiveness of intervention using probiotics to the detriment of other biotic agents in women's health, being bacterial vaginosis, polycystic ovary syndrome, and vulvovaginal candidiasis, the main diseases evaluated. However, preclinical studies have emphasized that the inhibition of pathogens responsible for the process of vaginal dysbiosis may be due to the formation of biofilm and the synthesis of compounds that could prevent the adhesion of these microorganisms. Future perspectives point to the beneficial modulation of the vaginal microbiota by biotic agents as a promising adjuvant approach to improve women's health.
Collapse
Affiliation(s)
- Douglas Xavier-Santos
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
- Fraunhofer Institute for Process Engineering and Packaging (IVV), Freising, Germany
| | - Raquel Bedani
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | | | - Marina Padilha
- Department of Social and Applied Nutrition, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Clara Mariana Gonçalves Lima
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Juliana Dara Rabêlo Silva
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Beatriz Manfrinato Ferreira
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Paulo César Giraldo
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Jorge Pamplona Pagnossa
- Department of Biological Sciences, Pontifical Catholic University, Poços de Caldas, MG, Brazil
| | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, SP, Brazil
| | | | - Anderson S Sant'Ana
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
8
|
Yi L, Mo A, Yang H, Yang Y, Xu Q, Yuan Y. Integrative RNA, miRNA, and 16S rRNA sequencing reveals immune-related regulation network for glycinin-induced enteritis in hybrid yellow catfish, Pelteobagrus fulvidraco ♀ × Pelteobagrus vachelli ♂. Front Immunol 2025; 15:1475195. [PMID: 39882244 PMCID: PMC11775447 DOI: 10.3389/fimmu.2024.1475195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Glycinin-induced foodborne enteritis is a significant obstacle that hinders the healthy development of the aquatic industry. Glycinin causes growth retardation and intestinal damage in hybrid yellow catfish (Pelteobagrus fulvidraco ♀ × Pelteobagrus vachelli ♂), but its immune mechanisms are largely unknown. In the current study, five experimental diets containing 0% (CK), 1.74% (G2), 3.57% (G4), 5.45% (G6), and 7.27% (G8) immunological activity of glycinin were fed to juvenile hybrid yellow catfish to reveal the mechanism of the intestinal immune response to glycinin through RNA and microRNA (miRNA) sequencing and to explore the interrelation between immune molecules and intestinal microbiota. The results demonstrated that glycinin content in the posterior intestine increased significantly and linearly with the rise of dietary glycinin levels. More than 5.45% of dietary glycinin significantly reduced the nutritional digestion and absorption function of the posterior intestine. Notably, an obvious alteration in the expression levels of inflammatory genes (tnf-α, il-1β, il-15, and tgf-β1) of the posterior intestine was observed when dietary glycinin exceeded 3.57%. Sequencing results of RNA and miRNA deciphered 4,246 differentially expressed genes (DEGs) and 28 differentially expressed miRNAs (DEmiRNAs) between the CK and G6 groups. Furthermore, enrichment analysis of DEGs and DEmiRNA target genes exhibited significant responses of the MAPK, NF-κB, and WNT pathways following experimental fish exposure to 5.45% dietary glycinin. Additionally, at the level of 3.57% in the diet, glycinin obviously inhibited the increase of microbiota, especially potential probiotics such as Ruminococcus bromii, Bacteroides plebeius, Faecalibacterium prausnitzii, and Clostridium clostridioforme. In sum, 5.45% dietary glycinin through the MAPK/NF-κB/WNT pathway induces enteritis, and inflammatory conditions could disrupt micro-ecological equilibrium through miRNA secreted by the host in hybrid yellow catfish. This study constitutes a comprehensive transcriptional perspective of how intestinal immunity responds to excessive glycinin in fish intestines.
Collapse
Affiliation(s)
- Linyuan Yi
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Aijie Mo
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huijun Yang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yifan Yang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qian Xu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yongchao Yuan
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei, China
- Shuangshui Shuanglu Institute, Huazhong Agricultural University, Wuhan, China
- National Demonstration Center for Experimental Aquaculture Education, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Liu Y, Wang F, Zhou Z, Liu B, Wu Z, Pan X. Profiling and comprehensive analysis of microbiome and ARGs of nurses and nursing workers in China: a cross-sectional study. Sci Rep 2024; 14:31301. [PMID: 39732868 PMCID: PMC11682234 DOI: 10.1038/s41598-024-82659-2] [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: 07/31/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Hospital-acquired infection (HAI) and antimicrobial resistance (AMR) represent major challenges in healthcare system. Despite numerous studies have assessed environmental and patient samples, very few studies have explored the microbiome and resistome profiles of medical staff including nursing workers. This cross-sectional study was performed in a tertiary hospital in China and involved 25 nurses (NSs), 25 nursing workers (NWs), and 55 non-medical control (NC). Stool samples from all participants and hand samples (i.e., the microbiome sample from hand skin, which were collected by swabbing both hands with a sponge-swab soaked with neutralized buffer and centrifuging the liquid buffer) from NSs and NWs were collected for metagenomic analysis. Metagenomic analysis revealed that medical staff exhibited lower abundances of beneficial species such as Blautia, and Bifidobacterium in the gut microbiome. However, an important potential pathogen, Staphylococcus haemolyticus, was enriched in the hands of NWs, suggesting a considerable prevalence of pathogenesis and multi-drug resistance. Accordantly, ARG analysis revealed worse hand hygiene among NWs than among NSs, characterized by a higher diversity of ARGs and a higher abundance of ARGs conferring multi-drug resistance including mdtF, acrB, AcrF and evgS. This study provides a comprehensive overview of the microbial and ARG profiles in the gut and hands of NSs and NWs. The higher abundance of potential pathogens and diverse multi-drug resistant ARGs in NWs hands indicates insufficient hand hygiene and a higher risk of HAI in this subgroup. This study is the first to highlight the critical need to improve hand hygiene among NWs, thus mitigating the risks of AMR and HAI.
Collapse
Affiliation(s)
- Ye Liu
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79, Qingchun Rd, Hangzhou, 310003, China
| | - Fang Wang
- Department of infectious diseases, Beilun District People's Hospital, Ningbo, 315800, Zhejiang, China
| | - Ziyuan Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Bowen Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhongwen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaxia Pan
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
10
|
Valentino V, De Filippis F, Marotta R, Pasolli E, Ercolini D. Genomic features and prevalence of Ruminococcus species in humans are associated with age, lifestyle, and disease. Cell Rep 2024; 43:115018. [PMID: 39615045 DOI: 10.1016/j.celrep.2024.115018] [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: 05/15/2024] [Revised: 09/23/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024] Open
Abstract
The genus Ruminococcus is dominant in the human gut, but higher levels of some species, such as R. gnavus, R. torques, and R. bromii, have been linked to health or disease. In this study, we analyzed >9,000 Ruminococcus metagenome-assembled genomes (MAGs) reconstructed from >5,000 subjects and revealed significant links between the prevalence of some species/subspecies and geographic origin, age, lifestyle, and disease, with subspecies prevalent in specific subpopulations showing divergent metabolic potential. Furthermore, Ruminococcus species from Lachnospiraceae encoded for carbohydrate-active enzymes (CAZy) potentially involved in the metabolism of human N- and O-glycans, whereas those from Oscillospiraceae appear to be more adapted toward fiber metabolism. These new findings contribute to elucidating the potential functional role of Ruminococcus in specific lifestyles and diseases and to decipher the diversity and the adaptation of members of this genus to the human gut.
Collapse
Affiliation(s)
- Vincenzo Valentino
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Roberto Marotta
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
11
|
Tiwari A, Ika Krisnawati D, Susilowati E, Mutalik C, Kuo TR. Next-Generation Probiotics and Chronic Diseases: A Review of Current Research and Future Directions. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27679-27700. [PMID: 39588716 DOI: 10.1021/acs.jafc.4c08702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The burgeoning field of microbiome research has profoundly reshaped our comprehension of human health, particularly highlighting the potential of probiotics and fecal microbiota transplantation (FMT) as therapeutic interventions. While the benefits of traditional probiotics are well-recognized, the efficacy and mechanisms remain ambiguous, and FMT's long-term effects are still being investigated. Recent advancements in high-throughput sequencing have identified gut microbes with significant health benefits, paving the way for next-generation probiotics (NGPs). These NGPs, engineered through synthetic biology and bioinformatics, are designed to address specific disease states with enhanced stability and viability. This review synthesizes current research on NGP stability, challenges in delivery, and their applications in preventing and treating chronic diseases such as diabetes, obesity, and cardiovascular diseases. We explore the physiological characteristics, safety profiles, and mechanisms of action of various NGP strains while also addressing the challenges and opportunities presented by their integration into clinical practice. The potential of NGPs to revolutionize microbiome-based therapies and improve clinical outcomes is immense, underscoring the need for further research to optimize their efficacy and ensure their safety.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Dyah Ika Krisnawati
- Department of Nursing, Faculty of Nursing and Midwifery, Universitas Nahdlatul Ulama Surabaya, Surabaya, 60237 East Java, Indonesia
| | - Erna Susilowati
- Akademi Kesehatan Dharma Husada Kediri, Kediri, 64118 East Java, Indonesia
| | - Chinmaya Mutalik
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-Rong Kuo
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
12
|
Lin TL, Chen WJ, Hung CM, Wong YL, Lu CC, Lai HC. Characterization and Safety Evaluation of Autoclaved Gut Commensal Parabacteroides goldsteinii RV-01. Int J Mol Sci 2024; 25:12660. [PMID: 39684372 DOI: 10.3390/ijms252312660] [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: 10/17/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Gut commensals play important roles in maintaining the homeostasis of human health. Previous studies indicated that the abundance of P. goldsteinii in animal hosts was increased by the administration of prebiotics such as polysaccharides purified from iconic oriental medicinal fungi. Subsequently, P. goldsteinii was found to exert beneficial effects on the amelioration of multiple chronic inflammation-associated diseases. Even so, during the process of the development of P. goldsteinii as a next-generation probiotic (NGP), care has to be taken when it is used as a functional food ingredient. In this study, we isolated a novel P. goldsteinii strain, RV-01, from the feces of a healthy adult and carried out comprehensive analyses of its genomic and phenotypic characteristics. Bioinformatic analysis of P. goldsteinii RV-01 revealed the absence of potential virulence genes, as well as the presence of genes and traits potentially beneficial to human health, such as the production of short-chain fatty acids, anti-inflammatory lipopolysaccharides, and zwitterionic capsular polysaccharides, as well as immune regulatory proteins. To circumvent any potential side effects, the P. goldsteinii RV-01 was autoclaved before proceeding to the nonclinical safety assessment. The autoclaved P. goldsteinii RV-01 retained its anti-inflammatory effect in human colon epithelial cells. In addition to the three genotoxicity assays, 28-day subacute and 90-day subchronic animal toxicity studies (the highest dose tested was equivalent to 8.109 × 1010P. goldsteinii RV-01 cells/kg body weight/day) were also implemented. The results of all studies were negative for toxicity. These results support the conclusion that autoclaved P. goldsteinii RV-01 is safe for use as a food ingredient.
Collapse
|
13
|
Naghibi M, Pont-Beltran A, Lamelas A, Llobregat L, Martinez-Blanch JF, Rojas A, Álvarez B, López Plaza B, Arcos Castellanos L, Chenoll E, Vijayakumar V, Day R. Effect of Postbiotic Bifidobacterium longum CECT 7347 on Gastrointestinal Symptoms, Serum Biochemistry, and Intestinal Microbiota in Healthy Adults: A Randomised, Parallel, Double-Blind, Placebo-Controlled Pilot Study. Nutrients 2024; 16:3952. [PMID: 39599737 PMCID: PMC11597252 DOI: 10.3390/nu16223952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVES A randomised, double-blind, placebo-controlled pilot trial was conducted to assess the effect of heat-treated Bifidobacterium longum CECT 7347 (HT-ES1) in healthy adults with mild to moderate digestive symptoms. A total of 60 participants were recruited and received either HT-ES1 or an identical placebo for 8 weeks with a further follow-up at week 10. METHODS This study monitored changes in the total Gastrointestinal Symptom Rating Scale for IBS score (GSRS-IBS), Irritable Bowel Syndrome Symptom Severity Scale (IBS-SSS), IBS Quality of Life index (IBS-QoL), gut microbiome using 16S rRNA sequencing, and the Visceral Sensitivity Index, as well as a range of biochemical markers, anthropometric parameters, and adverse events. RESULTS While minimal changes were observed in gastrointestinal (GI) symptoms, the HT-ES1 group showed a significant decrease in total and non-HDL cholesterol compared to the placebo. The intervention group also exhibited a significant increase in the abundance of the genera Faecalibacterium and Anaerobutyricum, both of which were positively correlated with butyrate concentrations. Faecal calprotectin significantly increased over time in the placebo group but remained stable in the HT-ES1 group. CONCLUSIONS Overall, these findings suggest that HT-ES1 may promote gut health by increasing butyrate-producing bacteria in the gut, maintaining normal levels of faecal calprotectin and reducing serum cholesterol.
Collapse
Affiliation(s)
- Malwina Naghibi
- Medical Department, ADM Health & Wellness, London SE1 7NT, UK
| | - Adria Pont-Beltran
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Araceli Lamelas
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Laura Llobregat
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Juan F. Martinez-Blanch
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Antonia Rojas
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Beatriz Álvarez
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | - Bricia López Plaza
- Food, Nutrition and Health Platform, Hospital La Paz Institzonulute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Medicine Department, Faculty of Medicine, Complutense University of Madrid, Plaza de Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - Lucia Arcos Castellanos
- Food, Nutrition and Health Platform, Hospital La Paz Institzonulute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Empar Chenoll
- ADM Research and Development Center-Valencia, ADM Health & Wellness, Parc Científic Universitat de València, 46980 València, Spain
| | | | - Richard Day
- Medical Department, ADM Health & Wellness, London SE1 7NT, UK
| |
Collapse
|
14
|
Jeyaraman M, Mariappan T, Jeyaraman N, Muthu S, Ramasubramanian S, Santos GS, da Fonseca LF, Lana JF. Gut microbiome: A revolution in type II diabetes mellitus. World J Diabetes 2024; 15:1874-1888. [PMID: 39280189 PMCID: PMC11372632 DOI: 10.4239/wjd.v15.i9.1874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/11/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Type II diabetes mellitus (T2DM) has experienced a dramatic increase globally across countries of various income levels over the past three decades. The persistent prevalence of T2DM is attributed to a complex interplay of genetic and environmental factors. While numerous pharmaceutical therapies have been developed, there remains an urgent need for innovative treatment approaches that offer effectiveness without significant adverse effects. In this context, the exploration of the gut microbiome presents a promising avenue. Research has increasingly shown that the gut microbiome of individuals with T2DM exhibits distinct differences compared to healthy individuals, suggesting its potential role in the disease's pathogenesis and progression. This emerging field offers diverse applications, particularly in modifying the gut environment through the administration of prebiotics, probiotics, and fecal microbiome transfer. These inter-ventions aim to restore a healthy microbiome balance, which could potentially alleviate or even reverse the metabolic dysfunctions associated with T2DM. Although current results from clinical trials have not yet shown dramatic effects on diabetes management, the groundwork has been laid for deeper investigation. Ongoing and future clinical trials are critical to advancing our understanding of the microbiome's impact on diabetes. By further elucidating the mechanisms through which microbiome alterations influence insulin resistance and glucose metabolism, researchers can develop more targeted interventions. The potential to harness the gut microbiome in developing new therapeutic strategies offers a compelling prospect to transform the treatment landscape of T2DM, potentially reducing the disease's burden significantly with approaches that are less reliant on traditional pharmaceuticals and more focused on holistic, systemic health improvements.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Tejaswin Mariappan
- Department of Community Medicine, Government Stanley Medical College and Hospital, Chennai 600001, Tamil Nadu, India
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Government Medical College, Karur 639004, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
15
|
Jan T, Negi R, Sharma B, Kumar S, Singh S, Rai AK, Shreaz S, Rustagi S, Chaudhary N, Kaur T, Kour D, Sheikh MA, Kumar K, Yadav AN, Ahmed N. Next generation probiotics for human health: An emerging perspective. Heliyon 2024; 10:e35980. [PMID: 39229543 PMCID: PMC11369468 DOI: 10.1016/j.heliyon.2024.e35980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024] Open
Abstract
Over recent years, the scientific community has acknowledged the crucial role of certain microbial strains inhabiting the intestinal ecosystem in promoting human health, and participating in various beneficial functions for the host. These microorganisms are now referred to as next-generation probiotics and are currently considered as biotherapeutic products and food or nutraceutical supplements. However, the majority of next-generation probiotic candidates pose nutritional demands and exhibit high sensitivity towards aerobic conditions, leading to numerous technological hurdles in large-scale production. This underscores the need for the development of suitable delivery systems capable of enhancing the viability and functionality of these probiotic strains. Currently, potential candidates for next generation probiotics (NGP) are being sought among gut bacteria linked to health, which include strains from the genera Bacteroids, Faecalibacterium, Akkermansia and Clostridium. In contrast to Lactobacillus spp. and Bifidobacterium spp., NGP, particularly Bacteroids spp. and Clostridium spp., appear to exhibit greater ambiguity regarding their potential to induce infectious diseases. The present review provides a comprehensive overview of NGPs in terms of their health beneficial effects, regulation framework and risk assessment targeting relevant criteria for commercialization in food and pharmaceutical markets.
Collapse
Affiliation(s)
- Tawseefa Jan
- Department of Food Technology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Rajeshwari Negi
- Department of Genetics, Plant Breeding and Biotechnology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Babita Sharma
- Department of Microbiology, Akal College of Basic Science, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Sanjeev Kumar
- Faculty of Agricultural Sciences, GLA University, Mathura, Uttar Pradesh, India
| | - Sangram Singh
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Ayodhya, Uttar Pradesh, India
| | - Ashutosh Kumar Rai
- Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Kingdom of Saudi Arabia
| | - Sheikh Shreaz
- Desert Agriculture and Ecosystem Department, Environment and Life Sciences Research Center, Kuwait Institute for Scientific Research, Safat, Kuwait
| | - Sarvesh Rustagi
- Depratment of Food Technology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Nisha Chaudhary
- Depratment of Food Science and Technology, Agriculture University, Jodhpur, Rajasthan, India
| | - Tanvir Kaur
- Department of Genetics, Plant Breeding and Biotechnology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Divjot Kour
- Department of Microbiology, Akal College of Basic Science, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Mohd Aaqib Sheikh
- Department of Food Technology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| | - Krishan Kumar
- Department of Food Technology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Ajar Nath Yadav
- Department of Genetics, Plant Breeding and Biotechnology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
- Chitkara Center for Research and Development, Chitkara University, Himachal Pradesh, India
| | - Naseer Ahmed
- Department of Food Technology, Dr. Khem Singh Gill Akal College of Agriculture, Eternal University, Baru Sahib, Sirmaur, Himachal Pradesh, India
| |
Collapse
|
16
|
Martínez A, Velázquez L, Díaz R, Huaiquipán R, Pérez I, Muñoz A, Valdés M, Sepúlveda N, Paz E, Quiñones J. Impact of Novel Foods on the Human Gut Microbiome: Current Status. Microorganisms 2024; 12:1750. [PMID: 39338424 PMCID: PMC11433882 DOI: 10.3390/microorganisms12091750] [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: 07/25/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
The microbiome is a complex ecosystem of microorganisms that inhabit a specific environment. It plays a significant role in human health, from food digestion to immune system strengthening. The "Novel Foods" refer to foods or ingredients that have not been consumed by humans in the European Union before 1997. Currently, there is growing interest in understanding how "Novel Foods" affect the microbiome and human health. The aim of this review was to assess the effects of "Novel Foods" on the human gut microbiome. Research was conducted using scientific databases, focusing on the literature published since 2000, with an emphasis on the past decade. In general, the benefits derived from this type of diet are due to the interaction between polyphenols, oligosaccharides, prebiotics, probiotics, fibre content, and the gut microbiome, which selectively promotes specific microbial species and increases microbial diversity. More research is being conducted on the consumption of novel foods to demonstrate how they affect the microbiome and, thus, human health. Consumption of novel foods with health-promoting properties should be further explored to maintain the diversity and functionality of the gut microbiome as a potential tool to prevent the onset and progression of chronic diseases.
Collapse
Affiliation(s)
- Ailín Martínez
- Doctoral Program in Science Major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4800000, Chile;
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
| | - Lidiana Velázquez
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Faculty of Agricultural and Environmental Sciences, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4780000, Chile;
| | - Rommy Díaz
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Faculty of Agricultural and Environmental Sciences, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4780000, Chile;
| | - Rodrigo Huaiquipán
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Doctoral Program in Agrifood and Environment Sciences, Universidad de La Frontera, Temuco 4780000, Chile
| | - Isabela Pérez
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Doctoral Program in Agrifood and Environment Sciences, Universidad de La Frontera, Temuco 4780000, Chile
| | - Alex Muñoz
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Doctoral Program in Agrifood and Environment Sciences, Universidad de La Frontera, Temuco 4780000, Chile
| | - Marcos Valdés
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Doctoral Program in Agrifood and Environment Sciences, Universidad de La Frontera, Temuco 4780000, Chile
| | - Néstor Sepúlveda
- Faculty of Agricultural and Environmental Sciences, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4780000, Chile;
- Doctoral Program in Agrifood and Environment Sciences, Universidad de La Frontera, Temuco 4780000, Chile
| | - Erwin Paz
- UWA Institute of Agriculture, The University of Western Australia, Perth 6009, Australia;
| | - John Quiñones
- Meat Quality Innovation and Technology Centre (CTI-Carne), Universidad de La Frontera, Temuco 4780000, Chile; (L.V.); (R.D.); (R.H.); (I.P.); (A.M.); (M.V.)
- Faculty of Agricultural and Environmental Sciences, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4780000, Chile;
| |
Collapse
|
17
|
Tain YL, Hou CY, Chang-Chien GP, Lin SF, Hsu CN. Chondroitin Sulfate Ameliorates Hypertension in Male Offspring Rat Born to Mothers Fed an Adenine Diet. Antioxidants (Basel) 2024; 13:944. [PMID: 39199190 PMCID: PMC11351932 DOI: 10.3390/antiox13080944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Pregnant women with chronic kidney disease (CKD) face increased risks of adverse outcomes in their adult offspring. Offspring rats born to dams fed an adenine diet develop hypertension, coinciding with dysregulated hydrogen sulfide (H2S) and nitric oxide (NO) pathways, as well as alterations in gut microbiota. Chondroitin sulfate (CS) is a multifunctional food known for its diverse bioactivities. As a sulfate prebiotic, CS has shown therapeutic potential in various diseases. Here, we investigated the protective effects of maternal CS supplementation against hypertension in offspring induced by an adenine diet. Mother rats were administered regular chow, 0.5% adenine, 3% CS, or a combination throughout gestation and lactation. Maternal CS supplementation effectively protected offspring from hypertension induced by the adenine diet. These beneficial effects of CS were connected with increased renal mRNA and protein levels of 3-mercaptopyruvate sulfurtransferase, an enzyme involved in H2S production. Furthermore, maternal CS treatment significantly enhanced alpha diversity and altered beta diversity of gut microbiota in adult offspring. Specifically, perinatal CS treatment promoted the abundance of beneficial microbes such as Roseburia hominis and Ruminococcus gauvreauii. In conclusion, perinatal CS treatment mitigates offspring hypertension associated with maternal adenine diet, suggesting that early administration of sulfate prebiotics may hold preventive potential. These findings warrant further translational research to explore their clinical implications.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 330, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan;
| | - Guo-Ping Chang-Chien
- Super Micro Mass Research and Technology Center, Cheng Shiu University, Kaohsiung 833, Taiwan; (G.-P.C.-C.); (S.-F.L.)
- Institute of Environmental Toxin and Emerging-Contaminant, Cheng Shiu University, Kaohsiung 833, Taiwan
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Shu-Fen Lin
- Super Micro Mass Research and Technology Center, Cheng Shiu University, Kaohsiung 833, Taiwan; (G.-P.C.-C.); (S.-F.L.)
- Institute of Environmental Toxin and Emerging-Contaminant, Cheng Shiu University, Kaohsiung 833, Taiwan
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
18
|
Trinh P, Teichman S, Roberts MC, Rabinowitz PM, Willis AD. A cross-sectional comparison of gut metagenomes between dairy workers and community controls. BMC Genomics 2024; 25:708. [PMID: 39033279 PMCID: PMC11626760 DOI: 10.1186/s12864-024-10562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND As a nexus of routine antibiotic use and zoonotic pathogen presence, the livestock farming environment is a potential hotspot for the emergence of zoonotic diseases and antibiotic resistant bacteria. Livestock can further facilitate disease transmission by serving as intermediary hosts for pathogens before a spillover event. In light of this, we aimed to characterize the microbiomes and resistomes of dairy workers, whose exposure to the livestock farming environment places them at risk for facilitating community transmission of antibiotic resistant genes and emerging zoonotic diseases. RESULTS Using shotgun sequencing, we investigated differences in the taxonomy, diversity and gene presence of 10 dairy farm workers and 6 community controls' gut metagenomes, contextualizing these samples with additional publicly available gut metagenomes. We found no significant differences in the prevalence of resistance genes, virulence factors, or taxonomic composition between the two groups. The lack of statistical significance may be attributed, in part, to the limited sample size of our study or the potential similarities in exposures between the dairy workers and community controls. We did, however, observe patterns warranting further investigation including greater abundance of tetracycline resistance genes and prevalence of cephamycin resistance genes as well as lower average gene diversity (even after accounting for differential sequencing depth) in dairy workers' metagenomes. We also found evidence of commensal organism association with tetracycline resistance genes in both groups (including Faecalibacterium prausnitzii, Ligilactobacillus animalis, and Simiaoa sunii). CONCLUSIONS This study highlights the utility of shotgun metagenomics in examining the microbiomes and resistomes of livestock workers, focusing on a cohort of dairy workers in the United States. While our study revealed no statistically significant differences between groups in taxonomy, diversity and gene presence, we observed patterns in antibiotic resistance gene abundance and prevalence that align with findings from previous studies of livestock workers in China and Europe. Our results lay the groundwork for future research involving larger cohorts of dairy and non-dairy workers to better understand the impact of occupational exposure to livestock farming on the microbiomes and resistomes of workers.
Collapse
Affiliation(s)
- Pauline Trinh
- Department of Biostatistics, University of Washington, Seattle, USA
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, USA
| | - Sarah Teichman
- Department of Statistics, University of Washington, Seattle, USA
| | - Marilyn C Roberts
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, USA
| | - Peter M Rabinowitz
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, USA
| | - Amy D Willis
- Department of Biostatistics, University of Washington, Seattle, USA.
- Department of Statistics, University of Washington, Seattle, USA.
| |
Collapse
|
19
|
Xing Y, Liu Y, Sha S, Zhang Y, Dou Y, Liu C, Xu M, Zhao L, Wang J, Wang Y, Ma X, Yan Q, Kong X. Multikingdom characterization of gut microbiota in patients with rheumatoid arthritis and rheumatoid arthritis-associated interstitial lung disease. J Med Virol 2024; 96:e29781. [PMID: 38961767 DOI: 10.1002/jmv.29781] [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: 01/13/2024] [Revised: 04/24/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a serious and common extra-articular disease manifestation. Patients with RA-ILD experience reduced bacterial diversity and gut bacteriome alterations. However, the gut mycobiome and virome in these patients have been largely neglected. In this study, we performed whole-metagenome shotgun sequencing on fecal samples from 30 patients with RA-ILD, and 30 with RA-non-ILD, and 40 matched healthy controls. The gut bacteriome and mycobiome were explored using a reference-based approach, while the gut virome was profiled based on a nonredundant viral operational taxonomic unit (vOTU) catalog. The results revealed significant alterations in the gut microbiomes of both RA-ILD and RA-non-ILD groups compared with healthy controls. These alterations encompassed changes in the relative abundances of 351 bacterial species, 65 fungal species, and 4,367 vOTUs. Bacteria such as Bifidobacterium longum, Dorea formicigenerans, and Collinsella aerofaciens were enriched in both patient groups. Ruminococcus gnavus (RA-ILD), Gemmiger formicilis, and Ruminococcus bromii (RA-non-ILD) were uniquely enriched. Conversely, Faecalibacterium prausnitzii, Bacteroides spp., and Roseburia inulinivorans showed depletion in both patient groups. Mycobiome analysis revealed depletion of certain fungi, including Saccharomyces cerevisiae and Candida albicans, in patients with RA compared with healthy subjects. Notably, gut virome alterations were characterized by an increase in Siphoviridae and a decrease in Myoviridae, Microviridae, and Autographiviridae in both patient groups. Hence, multikingdom gut microbial signatures showed promise as diagnostic indicators for both RA-ILD and RA-non-ILD. Overall, this study provides comprehensive insights into the fecal virome, bacteriome, and mycobiome landscapes of RA-ILD and RA-non-ILD gut microbiota, thereby offering potential biomarkers for further mechanistic and clinical research.
Collapse
Affiliation(s)
- Yida Xing
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yiping Liu
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shanshan Sha
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, China
| | - Yuemeng Dou
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Changyan Liu
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingxi Xu
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingdan Wang
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yan Wang
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiaochi Ma
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Qiulong Yan
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaodan Kong
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
20
|
Cattero V, Roussel C, Lessard-Lord J, Roy D, Desjardins Y. Supplementation with a cranberry extract favors the establishment of butyrogenic guilds in the human fermentation SHIME system. MICROBIOME RESEARCH REPORTS 2024; 3:34. [PMID: 39421251 PMCID: PMC11480733 DOI: 10.20517/mrr.2024.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 10/19/2024]
Abstract
Background: Proanthocyanidins (PAC) and oligosaccharides from cranberry exhibit multiple bioactive health properties and persist intact in the colon post-ingestion. They display a complex bidirectional interaction with the microbiome, which varies based on both time and specific regions of the gut; the nature of this interaction remains inadequately understood. Therefore, we aimed to investigate the impact of cranberry extract on gut microbiota ecology and function. Methods: We studied the effect of a cranberry extract on six healthy participants over a two-week supplementation period using the ex vivo artificial fermentation system TWIN-M-SHIME to replicate luminal and mucosal niches of the ascending and transverse colon. Results: Our findings revealed a significant influence of cranberry extract supplementation on the gut microbiota ecology under ex vivo conditions, leading to a considerable change in bacterial metabolism. Specifically, Bifidobacterium adolescentis (B. adolescentis) flourished in the mucus of the ascending colon, accompanied by a reduced adhesion of Proteobacteria. The overall bacterial metabolism shifted from acetate to propionate and, notably, butyrate production following PAC supplementation. Although there were variations in microbiota modulation among the six donors, the butyrogenic effect induced by the supplementation remained consistent across all individuals. This metabolic shift was associated with a rise in the relative abundance of several short-chain fatty acid (SCFA)-producing bacterial genera and the formation of a consortium of key butyrogenic bacteria in the mucus of the transverse colon. Conclusions: These observations suggest that cranberry extract supplementation has the potential to modulate the gut microbiota in a manner that may promote overall gut health.
Collapse
Affiliation(s)
- Valentina Cattero
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec City G1V 0A6, Quebec, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec City G1V 0A6, Quebec, Canada
| | - Charlène Roussel
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec City G1V 0A6, Quebec, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec City G1V 0A6, Quebec, Canada
- Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec City G1V 0A6, Quebec, Canada
| | - Jacob Lessard-Lord
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec City G1V 0A6, Quebec, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec City G1V 0A6, Quebec, Canada
| | - Denis Roy
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec City G1V 0A6, Quebec, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec City G1V 0A6, Quebec, Canada
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec City G1V 0A6, Quebec, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec City G1V 0A6, Quebec, Canada
| |
Collapse
|
21
|
Chai X, Chen X, Yan T, Zhao Q, Hu B, Jiang Z, Guo W, Zhang Y. Intestinal Barrier Impairment Induced by Gut Microbiome and Its Metabolites in School-Age Children with Zinc Deficiency. Nutrients 2024; 16:1289. [PMID: 38732540 PMCID: PMC11085614 DOI: 10.3390/nu16091289] [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: 03/10/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Zinc deficiency affects the physical and intellectual development of school-age children, while studies on the effects on intestinal microbes and metabolites in school-age children have not been reported. School-age children were enrolled to conduct anthropometric measurements and serum zinc and serum inflammatory factors detection, and children were divided into a zinc deficiency group (ZD) and control group (CK) based on the results of serum zinc. Stool samples were collected to conduct metagenome, metabolome, and diversity analysis, and species composition analysis, functional annotation, and correlation analysis were conducted to further explore the function and composition of the gut flora and metabolites of children with zinc deficiency. Beta-diversity analysis revealed a significantly different gut microbial community composition between ZD and CK groups. For instance, the relative abundances of Phocaeicola vulgatus, Alistipes putredinis, Bacteroides uniformis, Phocaeicola sp000434735, and Coprococcus eutactus were more enriched in the ZD group, while probiotic bacteria Bifidobacterium kashiwanohense showed the reverse trend. The functional profile of intestinal flora was also under the influence of zinc deficiency, as reflected by higher levels of various glycoside hydrolases in the ZD group. In addition, saccharin, the pro-inflammatory metabolites, and taurocholic acid, the potential factor inducing intestinal leakage, were higher in the ZD group. In conclusion, zinc deficiency may disturb the gut microbiome community and metabolic function profile of school-age children, potentially affecting human health.
Collapse
Affiliation(s)
- Xiaoqi Chai
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Xiaohui Chen
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Tenglong Yan
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Qian Zhao
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Binshuo Hu
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Zhongquan Jiang
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| | - Wei Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550000, China
| | - Ying Zhang
- School of Public Health, Lanzhou University, Lanzhou 730000, China; (X.C.); (X.C.); (T.Y.); (Q.Z.); (B.H.); (Z.J.)
| |
Collapse
|
22
|
Bedu-Ferrari C, Biscarrat P, Pepke F, Vati S, Chaudemanche C, Castelli F, Chollet C, Rué O, Hennequet-Antier C, Langella P, Cherbuy C. In-depth characterization of a selection of gut commensal bacteria reveals their functional capacities to metabolize dietary carbohydrates with prebiotic potential. mSystems 2024; 9:e0140123. [PMID: 38441031 PMCID: PMC11019791 DOI: 10.1128/msystems.01401-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/11/2024] [Indexed: 03/06/2024] Open
Abstract
The microbial utilization of dietary carbohydrates is closely linked to the pivotal role of the gut microbiome in human health. Inherent to the modulation of complex microbial communities, a prebiotic implies the selective utilization of a specific substrate, relying on the metabolic capacities of targeted microbes. In this study, we investigated the metabolic capacities of 17 commensal bacteria of the human gut microbiome toward dietary carbohydrates with prebiotic potential. First, in vitro experiments allowed the classification of bacterial growth and fermentation profiles in response to various carbon sources, including agave inulin, corn fiber, polydextrose, and citrus pectin. The influence of phylogenetic affiliation appeared to statistically outweigh carbon sources in determining the degree of carbohydrate utilization. Second, we narrowed our focus on six commensal bacteria representative of the Bacteroidetes and Firmicutes phyla to perform an untargeted high-resolution liquid chromatography-mass spectrometry metabolomic analysis: Bacteroides xylanisolvens, Bacteroides thetaiotaomicron, Bacteroides intestinalis, Subdoligranulum variabile, Roseburia intestinalis, and Eubacterium rectale exhibited distinct metabolomic profiles in response to different carbon sources. The relative abundance of bacterial metabolites was significantly influenced by dietary carbohydrates, with these effects being strain-specific and/or carbohydrate-specific. Particularly, the findings indicated an elevation in short-chain fatty acids and other metabolites, including succinate, gamma-aminobutyric acid, and nicotinic acid. These metabolites were associated with putative health benefits. Finally, an RNA-Seq transcriptomic approach provided deeper insights into the underlying mechanisms of carbohydrate metabolization. Restricting our focus on four commensal bacteria, including B. xylanisolvens, B. thetaiotaomicron, S. variabile, and R. intestinalis, carbon sources did significantly modulate the level of bacterial genes related to the enzymatic machinery involved in the metabolization of dietary carbohydrates. This study provides a holistic view of the molecular strategies induced during the dynamic interplay between dietary carbohydrates with prebiotic potential and gut commensal bacteria. IMPORTANCE This study explores at a molecular level the interactions between commensal health-relevant bacteria and dietary carbohydrates holding prebiotic potential. We showed that prebiotic breakdown involves the specific activation of gene expression related to carbohydrate metabolism. We also identified metabolites produced by each bacteria that are potentially related to our digestive health. The characterization of the functional activities of health-relevant bacteria toward prebiotic substances can yield a better application of prebiotics in clinical interventions and personalized nutrition. Overall, this study highlights the importance of identifying the impact of prebiotics at a low resolution of the gut microbiota to characterize the activities of targeted bacteria that can play a crucial role in our health.
Collapse
Affiliation(s)
- Cassandre Bedu-Ferrari
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
- General Mills France, Boulogne Billancourt, France
| | - Paul Biscarrat
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Frederic Pepke
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Sarah Vati
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Florence Castelli
- Service de Pharmacologie et Immuno-Analyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments,, CEA, INRAE, Université Paris Saclay, MetaboHUB, Gif-sur-Yvette, France
| | - Céline Chollet
- Service de Pharmacologie et Immuno-Analyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments,, CEA, INRAE, Université Paris Saclay, MetaboHUB, Gif-sur-Yvette, France
| | - Olivier Rué
- Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE bioinformatics facility, Jouy-en-Josas, France
| | - Christelle Hennequet-Antier
- Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE bioinformatics facility, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Claire Cherbuy
- Micalis Institute, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
23
|
Zwolschen JW, Vos AP, Ariëns RMC, Schols HA. In vitro batch fermentation of (un)saturated homogalacturonan oligosaccharides. Carbohydr Polym 2024; 329:121789. [PMID: 38286556 DOI: 10.1016/j.carbpol.2024.121789] [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: 10/23/2023] [Revised: 12/14/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Pectin, predominantly present within plant cell walls, is a dietary fiber that potentially induces distinct health effects depending on its molecular structure. Such structure-dependent health effects of pectin-derived galacturonic acid oligosaccharides (GalA-OS) are yet largely unknown. This study describes the influence of methyl-esterification and ∆4,5-unsaturation of GalA-OS through defined sets of GalA-OS made from pectin using defined pectinases, on the fermentability by individual fecal inocula. The metabolite production, OS utilization, quantity and size, methyl-esterification and saturation of remaining GalA-OS were monitored during the fermentation of GalA-OS. Fermentation of all GalA-OS predominantly induced the production of acetate, butyrate and propionate. Metabolization of unsaturated GalA-OS (uGalA-OS) significantly increased butyrate formation compared to saturated GalA-OS (satGalA-OS), while satGalA-OS significantly increased propionate formation. Absence of methyl-esters within GalA-OS improved substrate metabolization during the first 18 h of fermentation (99 %) compared to their esterified analogues (51 %). Furthermore, HPAEC and HILIC-LC-MS revealed accumulation of specific methyl-esterified GalA-OS, confirming that methyl-esterification delays fermentation. Fermentation of structurally distinct GalA-OS results in donor specific microbiota composition with uGalA-OS specifically stimulating the butyrate-producer Clostridium Butyricum. This study concludes that GalA-OS fermentation induces highly structure-dependent changes in the gut microbiota, further expanding their potential use as prebiotics.
Collapse
Affiliation(s)
- J W Zwolschen
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - A P Vos
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - R M C Ariëns
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - H A Schols
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands.
| |
Collapse
|
24
|
Cang W, Li X, Tang J, Wang Y, Mu D, Wu C, Shi H, Shi L, Wu J, Wu R. Therapeutic Potential of Bacteroides fragilis SNBF-1 as a Next-Generation Probiotic: In Vitro Efficacy in Lipid and Carbohydrate Metabolism and Antioxidant Activity. Foods 2024; 13:735. [PMID: 38472847 DOI: 10.3390/foods13050735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/11/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
This study explores the potential of aerotolerant Bacteroides fragilis (B. fragilis) strains as next-generation probiotics (NGPs), focusing on their adaptability in the gastrointestinal environment, safety profile, and probiotic functions. From 23 healthy infant fecal samples, we successfully isolated 56 beneficial B. fragilis strains. Notably, the SNBF-1 strain demonstrated superior cholesterol removal efficiency in HepG2 cells, outshining all other strains by achieving a remarkable reduction in cholesterol by 55.38 ± 2.26%. Comprehensive genotype and phenotype analyses were conducted, including sugar utilization and antibiotic sensitivity tests, leading to the development of an optimized growth medium for SNBF-1. SNBF-1 also demonstrated robust and consistent antioxidant activity, particularly in cell-free extracts, as evidenced by an average oxygen radical absorbance capacity value of 1.061 and a 2,2-diphenyl-1-picrylhydrazyl scavenging ability of 94.53 ± 7.31%. The regulation of carbohydrate metabolism by SNBF-1 was assessed in the insulin-resistant HepG2 cell line. In enzyme inhibition assays, SNBF-1 showed significant α-amylase and α-glucosidase inhibition, with rates of 87.04 ± 2.03% and 37.82 ± 1.36%, respectively. Furthermore, the cell-free supernatant (CFS) of SNBF-1 enhanced glucose consumption and glycogen synthesis in insulin-resistant HepG2 cells, indicating improved cellular energy metabolism. This was consistent with the observation that the CFS of SNBF-1 increased the proliferation of HepG2 cells by 123.77 ± 0.82% compared to that of the control. Overall, this research significantly enhances our understanding of NGPs and their potential therapeutic applications in modulating the gut microbiome.
Collapse
Affiliation(s)
- Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Xuan Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiayi Tang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Ying Wang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| | - Delun Mu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Chunting Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Haisu Shi
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Lin Shi
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, China
| |
Collapse
|
25
|
Calvete-Torre I, Sabater C, Cantabrana B, Margolles A, Sánchez M, Ruiz L. Gut microbiota modulation and effects of a diet enriched in apple pomace on inflammation in a DSS-induced colitis mouse model. Food Funct 2024; 15:2314-2326. [PMID: 38323473 DOI: 10.1039/d3fo04277d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Certain types of soluble dietary fibre, such as pectin and pectic oligosaccharides from different sources, have demonstrated protective effects against inflammation in DSS-induced colitis mouse models. In this work, we have evaluated the impact of a diet enriched in apple pomace (AP-diet), an agricultural by-product with a significant content of pectin and that previously demonstrated prebiotic properties in human fecal batch fermentation models, on the gut microbiota composition, intestinal damage and inflammation markers in a DSS-induced colitis model. We found that the apple pomace enriched diet (AP-diet), providing a significant amount of pectin with demonstrated prebiotic properties, was associated with a slower increase in the disease activity index, translating into better clinical symptomatology of the animals. Histological damage scoring confirmed less severe damage in those animals receiving an AP-diet before and during the DSS administration period. Some serum inflammatory markers, such as TNFα, also demonstrated lower levels in the group receiving the AP-diet, compared to the control diet. AP-diet administration is also associated with the modulation of key taxa in the colonic microbiota of animals, such as some Lachnospiraceae genera and Ruminococcus species, including commensal short chain fatty acid producers that could play a role in attenuating inflammation at the intestinal level.
Collapse
Affiliation(s)
- Inés Calvete-Torre
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Begoña Cantabrana
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Pharmacology of Therapeutic Targets Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Manuel Sánchez
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Pharmacology of Therapeutic Targets Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.
- Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| |
Collapse
|
26
|
Li L, Liu T, Shi Y. Treatment of preterm brain injury via gut-microbiota-metabolite-brain axis. CNS Neurosci Ther 2024; 30:e14556. [PMID: 38108213 PMCID: PMC10805406 DOI: 10.1111/cns.14556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Brain injury in preterm infants potentially disrupts critical structural and functional connective networks in the brain. It is a major cause of neurological sequelae and developmental deficits in preterm infants. Interesting findings suggest that the gut microbiota (GM) and their metabolites contribute to the programming of the central nervous system (CNS) during developmental stages and may exert structural and functional effects throughout the lifespan. AIM To summarize the existing knowledge of the potential mechanisms related to immune, endocrine, neural, and blood-brain barrier (BBB) mediated by GM and its metabolites in neural development and function. METHODS We review the recent literature and included 150 articles to summarize the mechanisms through which GM and their metabolites work on the nervous system. Potential health benefits and challenges of relevant treatments are also discussed. RESULTS This review discusses the direct and indirect ways through which the GM may act on the nervous system. Treatment of preterm brain injury with GM or related derivatives, including probiotics, prebiotics, synbiotics, dietary interventions, and fecal transplants are also included. CONCLUSION This review summarizes mechanisms underlying microbiota-gut-brain axis and novel therapeutic opportunities for neurological sequelae in preterm infants. Optimizing the initial colonization and microbiota development in preterm infants may represent a novel therapy to promote brain development and reduce long-term sequelae.
Collapse
Affiliation(s)
- Ling Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Tianjing Liu
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Yongyan Shi
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
27
|
Ngom SI, Maski S, Rached B, Chouati T, Oliveira Correia L, Juste C, Meylheuc T, Henrissat B, El Fahime E, Amar M, Béra-Maillet C. Exploring the hemicellulolytic properties and safety of Bacillus paralicheniformis as stepping stone in the use of new fibrolytic beneficial microbes. Sci Rep 2023; 13:22785. [PMID: 38129471 PMCID: PMC10740013 DOI: 10.1038/s41598-023-49724-8] [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: 01/20/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Bacillus strains from the Moroccan Coordinated Collections of Microorganisms (CCMM) were characterised and tested for fibrolytic function and safety properties that would be beneficial for maintaining intestinal homeostasis, and recommend beneficial microbes in the field of health promotion research. Forty strains were investigated for their fibrolytic activities towards complex purified polysaccharides and natural fibres representative of dietary fibres (DFs) entering the colon for digestion. We demonstrated hemicellulolytic activities for nine strains of Bacillus aerius, re-identified as Bacillus paralicheniformis and Bacillus licheniformis, using xylan, xyloglucan or lichenan as purified polysaccharides, and orange, apple and carrot natural fibres, with strain- and substrate-dependent production of glycoside hydrolases (GHs). Our combined methods, based on enzymatic assays, secretome, and genome analyses, highlighted the hemicellulolytic activities of B. paralicheniformis and the secretion of specific glycoside hydrolases, in particular xylanases, compared to B. licheniformis. Genomic features of these strains revealed a complete set of GH genes dedicated to the degradation of various polysaccharides from DFs, including cellulose, hemicellulose and pectin, which may confer on the strains the ability to digest a variety of DFs. Preliminary experiments on the safety and immunomodulatory properties of B. paralicheniformis fibrolytic strains were evaluated in light of applications as beneficial microbes' candidates for health improvement. B. paralicheniformis CCMM B969 was therefore proposed as a new fibrolytic beneficial microbe candidate.
Collapse
Affiliation(s)
- Serigne Inssa Ngom
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Soufiane Maski
- Laboratoire de Microbiologie et Biologie Moléculaire, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Département de Biologie, Faculté des Sciences, Université Mohammed V, Rabat, Morocco
| | - Bahia Rached
- Collections Coordonnées Marocaines de Microorganismes, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Plateforme Génomique Fonctionnelle, Unité d'Appui Technique à la Recherche Scientifique, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Laboratoire de Chimie-Physique et Biotechnologies des Biomolécules et Matériaux/Equipe Microbiologie Biomolécules et Biotechnologies, Faculté des Sciences et Techniques, Mohammedia, Morocco
| | - Taha Chouati
- Collections Coordonnées Marocaines de Microorganismes, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Plateforme Génomique Fonctionnelle, Unité d'Appui Technique à la Recherche Scientifique, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Biologie médicale, Pathologie humaine et Expérimentale et Environnement, Faculté de Médecine et de pharmacie de Rabat, Rabat, Morocco
| | - Lydie Oliveira Correia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, PAPPSO, 78350, Jouy-en-Josas, France
| | - Catherine Juste
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Thierry Meylheuc
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, MIMA2, 78350, Jouy en Josas, France
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique, Aix-Marseille Université, 13288, Marseille, France
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Elmostafa El Fahime
- Plateforme Génomique Fonctionnelle, Unité d'Appui Technique à la Recherche Scientifique, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Biologie médicale, Pathologie humaine et Expérimentale et Environnement, Faculté de Médecine et de pharmacie de Rabat, Rabat, Morocco
| | - Mohamed Amar
- Laboratoire de Microbiologie et Biologie Moléculaire, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
- Collections Coordonnées Marocaines de Microorganismes, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco
| | - Christel Béra-Maillet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France.
- Laboratoire de Microbiologie et Biologie Moléculaire, Centre National pour la Recherche Scientifique et Technique, Rabat, Morocco.
| |
Collapse
|
28
|
Avelar-Barragan J, Mendez Luque LF, Nguyen J, Nguyen H, Odegaard AO, Fleischman AG, Whiteson KL. Characterizing the microbiome of patients with myeloproliferative neoplasms during a Mediterranean diet intervention. mBio 2023; 14:e0230823. [PMID: 37877698 PMCID: PMC10746218 DOI: 10.1128/mbio.02308-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
IMPORTANCE The gut microbiome serves as an interface between the host and the diet. Diet and the gut microbiome both play important roles in managing inflammation, which is a key aspect of myeloproliferative neoplasm (MPN). Studies have shown that a Mediterranean (MED) diet can reduce inflammation. Therefore, we longitudinally characterized the gut microbiomes of MPN patients in response to Mediterranean or standard 2020 US Guidelines for Americans dietary counseling to determine whether there were microbiome-associated changes in inflammation. We did not find significant changes in the gut microbiome associated with diet, but we did find several associations with inflammation. This research paves the way for future studies by identifying potential mechanistic targets implicated in inflammation within the MPN gut microbiome.
Collapse
Affiliation(s)
- Julio Avelar-Barragan
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA
| | - Laura F. Mendez Luque
- Department of Biological Chemistry, University of California Irvine, Irvine, California, USA
| | - Jenny Nguyen
- Division of Hematology/Oncology, University of California Irvine, Irvine, California, USA
| | - Hellen Nguyen
- Division of Hematology/Oncology, University of California Irvine, Irvine, California, USA
| | - Andrew O. Odegaard
- Department of Epidemiology and Biostatistics, University of California Irvine, Irvine, California, USA
| | - Angela G. Fleischman
- Department of Biological Chemistry, University of California Irvine, Irvine, California, USA
- Division of Hematology/Oncology, University of California Irvine, Irvine, California, USA
| | - Katrine L. Whiteson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA
| |
Collapse
|
29
|
Lakshmanan AP, Deola S, Terranegra A. The Promise of Precision Nutrition for Modulation of the Gut Microbiota as a Novel Therapeutic Approach to Acute Graft-versus-host Disease. Transplantation 2023; 107:2497-2509. [PMID: 37189240 PMCID: PMC10664798 DOI: 10.1097/tp.0000000000004629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/08/2023] [Accepted: 02/20/2023] [Indexed: 05/17/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a severe side effect of allogeneic hematopoietic stem cell transplantation (aHSCT) that has complex phenotypes and often unpredictable outcomes. The current management is not always able to prevent aGVHD. A neglected actor in the management of aGVHD is the gut microbiota. Gut microbiota dysbiosis after aHSCT is caused by many factors and may contribute to the development of aGVHD. Diet and nutritional status modify the gut microbiota and a wide range of products are now available to manipulate the gut microbiota (pro-, pre-, and postbiotics). New investigations are testing the effect of probiotics and nutritional supplements in both animal models and human studies, with encouraging results. In this review, we summarize the most recent literature about the probiotics and nutritional factors able to modulate the gut microbiota and we discuss the future perspective in developing new integrative therapeutic approaches to reducing the risk of graft-versus-host disease in patients undergoing aHSCT.
Collapse
Affiliation(s)
| | - Sara Deola
- Advanced Cell Therapy Core, Research Branch, Sidra Medicine, Qatar
| | | |
Collapse
|
30
|
Choi SP, Park SW, Kang SJ, Lim SK, Kwon MS, Choi HJ, Chun T. Monitoring mRNA Expression Patterns in Macrophages in Response to Two Different Strains of Probiotics. Food Sci Anim Resour 2023; 43:703-711. [PMID: 37484002 PMCID: PMC10359838 DOI: 10.5851/kosfa.2023.e23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 07/25/2023] Open
Abstract
As an initial study to elucidate the molecular mechanism of how probiotics modulate macrophage activity, we monitored mRNA expression patterns in peritoneal macrophages (PMs) treated with two different strains of probiotics. After treatment with either Weissella cibaria WIKIM28 or Latilactobacillus sakei WIKIM50, total RNAs from PMs were isolated and subjected into gene chip analyses. As controls, mRNAs from vehicle (phosphate-buffered saline, PBS)-treated PMs were also subjected to gene chip analysis. Compared to vehicle (PBS)-treated PMs, WIKIM28-treated and WIKIM50-treated PMs exhibited a total of 889 and 432 differentially expressed genes with expression differences of at least 4 folds, respectively. Compared to WIKIM28-treated PMs, WIKIM50-treated PMs showed 25 up-regulated genes and 21 down-regulated genes with expression differences of more than 2 folds. Interestingly, mRNA transcripts of M2 macrophage polarization marker such as anxa1, mafb, and sepp1 were increased in WIKIM50-treated PMs comparing to those in WIKIM28-treated PMs. Reversely, mRNA transcripts of M1 macrophage polarization marker such as hdac9, ptgs2, and socs3 were decreased in WIKIM50-treated PMs comparing to those in WIKIM28-treated PMs. In agreement with these observations, mRNA expression levels of tumor necrosis factor-α and interleukin-1α were significantly reduced in WIKIM50-treated macrophages compared to those in WIKIM28-treated macrophages. These results may indicate that probiotics can be classified as two different types depending on their ability to convert macrophages into M1 or M2 polarization.
Collapse
Affiliation(s)
- Sang-Pil Choi
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Si-Won Park
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Seok-Jin Kang
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Seul Ki Lim
- Technology Innovation Research Division,
World Institute of Kimchi, Gwangju 61755, Korea
| | - Min-Sung Kwon
- Technology Innovation Research Division,
World Institute of Kimchi, Gwangju 61755, Korea
| | - Hak-Jong Choi
- Technology Innovation Research Division,
World Institute of Kimchi, Gwangju 61755, Korea
| | - Taehoon Chun
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| |
Collapse
|
31
|
Chiantera V, Laganà AS, Basciani S, Nordio M, Bizzarri M. A Critical Perspective on the Supplementation of Akkermansia muciniphila: Benefits and Harms. Life (Basel) 2023; 13:1247. [PMID: 37374030 DOI: 10.3390/life13061247] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Akkermansia muciniphila is a mucin-degrading bacterium of the intestinal niche, exerting beneficial effects on the host metabolic profile. Accumulating evidence indicated Akkermansia as a promising therapeutic probiotic against metabolic disorders such as obesity, type 2 diabetes and cardiovascular diseases. However, in specific intestinal microenvironments, its excessive enrichment may be not beneficial. Conditions like inflammatory bowel disease (IBD), Salmonella typhimurium infection or post-antibiotic reconstitution may not benefit from Akkermansia supplementation. Furthermore, using Akkermansia in patients with endocrine and gynecological disorders-such as polycystic ovary syndrome (PCOS) or endometriosis-that have a higher risk of developing IBD, should be critically evaluated. In addition, a cautionary note comes from the neurological field, as the gut microbiota of patients suffering from Parkinson's disease or multiple sclerosis exhibits a characteristic signature of Akkermansia municiphila abundance. Overall, considering these controversial points, the use of Akkermansia should be evaluated on an individual basis, avoiding risking unexpected effects.
Collapse
Affiliation(s)
- Vito Chiantera
- Unit of Gynecologic Oncology, ARNAS "Civico-Di Cristina-Benfratelli", Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS "Civico-Di Cristina-Benfratelli", Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Sabrina Basciani
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maurizio Nordio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Mariano Bizzarri
- System Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| |
Collapse
|
32
|
Trinh P, Roberts MC, Rabinowitz PM, Willis AD. Differences in gut metagenomes between dairy workers and community controls: a cross-sectional study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540270. [PMID: 37215025 PMCID: PMC10197731 DOI: 10.1101/2023.05.10.540270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background As a nexus of routine antibiotic use and zoonotic pathogen presence, the livestock farming environment is a potential hotspot for the emergence of zoonotic diseases and antibiotic resistant bacteria. Livestock can further facilitate disease transmission by serving as intermediary hosts for pathogens as they undergo evolution prior to a spillover event. In light of this, we are interested in characterizing the microbiome and resistome of dairy workers, whose exposure to the livestock farming environment places them at risk for facilitating community transmission of antibiotic resistant genes and emerging zoonotic diseases. Results Using shotgun sequencing, we investigated differences in the taxonomy, diversity and gene presence of the human gut microbiome of 10 dairy farm workers and 6 community controls, supplementing these samples with additional publicly available gut metagenomes. We observed greater abundance of tetracycline resistance genes and prevalence of cephamycin resistance genes in dairy workers' metagenomes, and lower average gene diversity. We also found evidence of commensal organism association with plasmid-mediated tetracycline resistance genes in both dairy workers and community controls (including Faecalibacterium prausnitzii, Ligilactobacillus animalis, and Simiaoa sunii). However, we did not find significant differences in the prevalence of resistance genes or virulence factors overall, nor differences in the taxonomic composition of dairy worker and community control metagenomes. Conclusions This study presents the first metagenomics analysis of United States dairy workers, providing insights into potential risks of exposure to antibiotics and pathogens in animal farming environments. Previous metagenomic studies of livestock workers in China and Europe have reported increased abundance and carriage of antibiotic resistance genes in livestock workers. While our investigation found no strong evidence for differences in the abundance or carriage of antibiotic resistance genes and virulence factors between dairy worker and community control gut metagenomes, we did observe patterns in the abundance of tetracycline resistance genes and the prevalence of cephamycin resistance genes that is consistent with previous work.
Collapse
Affiliation(s)
- Pauline Trinh
- Department of Environmental & Occupational Health Sciences, University of Washington
- Department of Biostatistics, University of Washington
| | - Marilyn C Roberts
- Department of Environmental & Occupational Health Sciences, University of Washington
| | - Peter M Rabinowitz
- Department of Environmental & Occupational Health Sciences, University of Washington
| | - Amy D Willis
- Department of Biostatistics, University of Washington
| |
Collapse
|
33
|
Pellegrino A, Coppola G, Santopaolo F, Gasbarrini A, Ponziani FR. Role of Akkermansia in Human Diseases: From Causation to Therapeutic Properties. Nutrients 2023; 15:nu15081815. [PMID: 37111034 PMCID: PMC10142179 DOI: 10.3390/nu15081815] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The gut microbiota plays a critical role in the modulation of host metabolism and immune response, and its impairment has been implicated in many gastrointestinal and extraintestinal diseases. Current evidence shows the well-documented role of A. muciniphila in maintaining the integrity of the intestinal barrier, modulating the host immune response, and improving several metabolic pathways, making it a key element in the pathogenesis of several human diseases. In this scenario, A. muciniphila is the most promising next-generation probiotic and one of the first microbial species suitable for specific clinical use when compared with traditional probiotics. Further studies are needed to provide more accurate insight into its mechanisms of action and to better elucidate its properties in several major areas, paving the way for a more integrated and personalized therapeutic approach that finally makes the most of our knowledge of the gut microbiota.
Collapse
Affiliation(s)
- Antonio Pellegrino
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Gaetano Coppola
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
34
|
Xie Z, Zhang G, Liu R, Wang Y, Tsapieva AN, Zhang L, Han J. Heat-Killed Lacticaseibacillus paracasei Repairs Lipopolysaccharide-Induced Intestinal Epithelial Barrier Damage via MLCK/MLC Pathway Activation. Nutrients 2023; 15:nu15071758. [PMID: 37049598 PMCID: PMC10097264 DOI: 10.3390/nu15071758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Intestinal epithelial barrier function is closely associated with the development of many intestinal diseases. Heat-killed Lacticaseibacillus paracasei (HK-LP) has been shown to improve intestinal health and enhance immunity. However, the function of HK-LP in the intestinal barrier is still unclear. This study characterized the inflammatory effects of seven HK-LP (1 μg/mL) on the intestinal barrier using lipopolysaccharide (LPS) (100 μg/mL)-induced Caco-2 cells. In this study, HK-LP 6105, 6115, and 6235 were selected, and their effects on the modulation of inflammatory factors and tight junction protein expression (claudin-1, zona occludens-1, and occludin) were compared. The effect of different cultivation times (18 and 48 h) was investigated in response to LPS-induced intestinal epithelial barrier dysfunction. Our results showed that HK-LP 6105, 6115, and 6235 improved LPS-induced intestinal barrier permeability reduction and transepithelial resistance. Furthermore, HK-LP 6105, 6115, and 6235 inhibited the pro-inflammatory factors (TNF-α, IL-1β, IL-6) and increased the expression of the anti-inflammatory factors (IL-4, IL-10, and TGF-β). HK-LP 6105, 6115, and 6235 ameliorated the inflammatory response. It inhibited the nuclear factor kappa B (NF-κB) signaling pathway-mediated myosin light chain (MLC)/MLC kinase signaling pathway by downregulating the Toll-like receptor 4 (TLR4)/NF-κB pathway. Thus, the results suggest that HK-LP 6150, 6115, and 6235 may improve intestinal health by regulating inflammation and TJ proteins. Postbiotics produced by these strains exhibit anti-inflammatory properties that can protect the intestinal barrier.
Collapse
Affiliation(s)
- Zhixin Xie
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Gongsheng Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Rongxu Liu
- Heilongjiang Green Food Science Research Institute, Harbin 150030, China
| | - Yucong Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Anna N Tsapieva
- Department of Molecular Microbiology, FSBSI Institute of Experimental Medicine, Acad.,197376 St. Petersburg, Russia
| | - Lili Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jianchun Han
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Green Food Science Research Institute, Harbin 150030, China
| |
Collapse
|
35
|
Chen C, Zhang Y, Yao X, Li S, Wang G, Huang Y, Yang Y, Zhang A, Liu C, Zhu D, Li H, Yan Q, Ma W. Characterizations of the Gut Bacteriome, Mycobiome, and Virome in Patients with Osteoarthritis. Microbiol Spectr 2023; 11:e0171122. [PMID: 36515546 PMCID: PMC9927108 DOI: 10.1128/spectrum.01711-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota plays an essential role in the regulation of the immune system and the etiology of human autoimmune diseases. However, a holistic understanding of the gut bacteriome, mycobiome, and virome in patients with osteoarthritis (OA) remains lacking. Here, we explored the gut microbiotas of 44 OA patients and 46 healthy volunteers via deep whole-metagenome shotgun sequencing of their fecal samples. The gut bacteriome and mycobiome were analyzed using a reference-based strategy. Gut viruses were identified from the metagenomic assembled contigs, and the gut virome was profiled based on 6,567 nonredundant viral operational taxonomic units (vOTUs). We revealed that the gut microbiome (including bacteriome, mycobiome, and virome) of OA patients is fundamentally altered, characterized by a panel of 279 differentially abundant bacterial species, 10 fungal species, and 627 vOTUs. The representative OA-enriched bacteria included Anaerostipes hadrus (GENOME147149), Prevotella sp900313215 (GENOME08259), Eubacterium_E hallii (GENOME000299), and Blautia A (GENOME001004), while Bacteroides plebeius A (GENOME239725), Roseburia inulinivorans (GENOME 001770), Dialister sp900343095 (GENOME075103), Phascolarctobacterium faecium (GENOME233517), and several members of Faecalibacterium and Prevotella were depleted in OA patients. Fungi such as Debaryomyces fabryi (GenBank accession no. GCA_003708665), Candida parapsilosis (GCA_000182765), and Apophysomyces trapeziformis (GCA_000696975) were enriched in the OA gut microbiota, and Malassezia restricta (GCA_003290485), Aspergillus fumigatus (GCA_003069565), and Mucor circinelloides (GCA_010203745) were depleted. The OA-depleted viruses spanned Siphoviridae (95 vOTUs), Myoviridae (70 vOTUs), and Microviridae (5 vOTUs), while 30 Siphoviridae vOTUs were enriched in OA patients. Functional analysis of the gut bacteriome and virome also uncovered their functional signatures in relation to OA. Moreover, we demonstrated that the OA-associated gut bacterial and viral signatures are tightly interconnected, suggesting that they may impact disease together. Finally, we showed that the multikingdom signatures are effective in discriminating the OA patients from healthy controls, suggesting the potential of gut microbiota for the prediction of OA and related diseases. Our results delineated the fecal bacteriome, mycobiome, and virome landscapes of the OA microbiota and provided biomarkers that will aid in future mechanistic and clinical intervention studies. IMPORTANCE The gut microbiome of OA patients was completely altered compared to that in healthy individuals, including 279 differentially abundant bacterial species, 10 fungal species and 627 viral operational taxonomic units (vOTUs). Functional analysis of the gut bacteriome and virome also revealed their functional signatures in relation to OA. We found that OA-associated gut bacterial and viral signatures were tightly interconnected, indicating that they may affect the disease together. The OA patients can be discriminated effectively from healthy controls using the multikingdom signatures, suggesting the potential of gut microbiota for the prediction of OA and related diseases.
Collapse
Affiliation(s)
- Changming Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, China
| | - Xueming Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | | | - Guangyang Wang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yutao Yang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | | | - Can Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dan Zhu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hufan Li
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiulong Yan
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Wukai Ma
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
36
|
Begum N, Mandhare A, Tryphena KP, Srivastava S, Shaikh MF, Singh SB, Khatri DK. Epigenetics in depression and gut-brain axis: A molecular crosstalk. Front Aging Neurosci 2022; 14:1048333. [PMID: 36583185 PMCID: PMC9794020 DOI: 10.3389/fnagi.2022.1048333] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Gut-brain axis is a dynamic, complex, and bidirectional communication network between the gut and brain. Changes in the microbiota-gut-brain axis are responsible for developing various metabolic, neurodegenerative, and neuropsychiatric disorders. According to clinical and preclinical findings, the gut microbiota is a significant regulator of the gut-brain axis. In addition to interacting with intestinal cells and the enteric nervous system, it has been discovered that microbes in the gut can modify the central nervous system through metabolic and neuroendocrine pathways. The metabolites of the gut microbiome can modulate a number of diseases by inducing epigenetic alteration through DNA methylation, histone modification, and non-coding RNA-associated gene silencing. Short-chain fatty acids, especially butyrate, are well-known histone deacetylases inhibitors. Similarly, other microbial metabolites such as folate, choline, and trimethylamine-N-oxide also regulate epigenetics mechanisms. Furthermore, various studies have revealed the potential role of microbiome dysbiosis and epigenetics in the pathophysiology of depression. Hence, in this review, we have highlighted the role of gut dysbiosis in epigenetic regulation, causal interaction between host epigenetic modification and the gut microbiome in depression and suggest microbiome and epigenome as a possible target for diagnosis, prevention, and treatment of depression.
Collapse
Affiliation(s)
- Nusrat Begum
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Aniket Mandhare
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Kamatham Pushpa Tryphena
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India,*Correspondence: Saurabh Srivastava,
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia,Mohd Farooq Shaikh,
| | - Shashi Bala Singh
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India,Dharmendra Kumar Khatri,
| |
Collapse
|
37
|
Rani K, Ali SA, Kaul G, Behare PV. Protective effect of probiotic and prebiotic fermented milk containing Lactobacillus fermentum against obesity-induced hepatic steatosis and inflammation. J Food Biochem 2022; 46:e14509. [PMID: 36334279 DOI: 10.1111/jfbc.14509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/08/2022]
Abstract
Obesity has reached epidemic proportions, with major economic and health implications. The complex pathophysiology of obesity explains the difficulty provided to health policy for its clinical management. Increasing data show that obesity and metabolic abnormalities are intimately connected to differences in consumption of probiotics, its relevance to gut microbiota activity and composition. The goal of this investigation was to assess the effect of oral delivery of indigenous probiotic Lactobacillus fermentum NCDC 400 and prebiotic fructo-oligosaccharide (FOS) on obesity-associated hepatic steatosis and inflammation produced by a high-fat diet (HFD). C57BL/6 mice treated with L. fermentum NCDC 400 either independently or in conjunction with FOS demonstrated reduced body weight and abdominal obesity after 24 weeks of treatment. Also, the anti-oxidative enzyme activity went down, and the inflammatory profile got better, with less fat getting into the hepatocytes. The lipid profile changed, with HDL cholesterol going up and LDL cholesterol and triglyceride levels going down. Further, L. fermentum NCDC 400 and FOS combinations decreased fasting glucose, gHbA1c, gastric inhibitory peptide, and insulin levels in mice fed with HFD, thus improving glucose homeostasis. Overall, consumption of L. fermentum NCDC 400 alone or its combinational effects had a protective role on obesity-associated hepatic steatosis. PRACTICAL APPLICATIONS: The potential indigenous probiotic Lactobacillus fermentum NCDC 400 and prebiotic FOS had a preventive role in obesity-induced hepatic steatosis and improves anti-oxidant and anti-inflammatory properties in HFD-fed obese mice. Our finding would be helpful to prevent obesity-associated hepatic steatosis and inflammation upon supplementation of pre- and pro-biotics (synbiotics).
Collapse
Affiliation(s)
- Kavita Rani
- Semen Biology Lab, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Haryana, India
| | - Syed Azmal Ali
- German Cancer Research Center, Division Proteomics of Stem Cells and Cancer, Heidelberg, Germany.,Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| | - Gautam Kaul
- Semen Biology Lab, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Haryana, India
| | - Pradip V Behare
- National Collection of Dairy Cultures (NCDC) Lab, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
38
|
Filardi R, Gargari G, Mora D, Arioli S. Characterization of antibiotic-resistance traits in Akkermansia muciniphila strains of human origin. Sci Rep 2022; 12:19426. [PMID: 36371559 PMCID: PMC9653403 DOI: 10.1038/s41598-022-23980-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
Akkermansia muciniphila, a commensal bacterium commonly found in healthy gut microbiota, is widely considered a next-generation beneficial bacterium candidate to improve metabolic and inflammatory disorders. Recently the EFSA's Panel on Nutrition, Novel food, and Food Allergens has declared that pasteurized A. muciniphila DSM 22959T (also MucT, ATCC BAA-835) can be considered safe as a novel food, opening the door to its commercialization as a food supplement. Despite its recognized health benefits, there is still little information regarding the antimicrobial susceptibility of this species and reference cut-off values to distinguish strains with intrinsic or acquired resistance from susceptible strains. In this study, we combined a genomic approach with the evaluation of the antibiotic susceptibility in five human A. muciniphila isolates. Genomic mining for antimicrobial resistance genes and MICs determinations revealed that only one strain harboring tetW gene showed resistance to tetracycline, whereas all A. muciniphila strains showed low sensitivity to ciprofloxacin and aminoglycosides with no genotypic correlation. Although all strains harbor the gene adeF, encoding for a subunit of the resistance-nodulation-cell division efflux pump system, potentially involved in ciprofloxacin resistance, the susceptibility towards ciprofloxacin determined in presence of efflux pump inhibitors was not affected. Overall, our outcomes revealed the importance to extend the antibiotic susceptibility test to a larger number of new isolates of A. muciniphila to better assess the safety aspects of this species.
Collapse
Affiliation(s)
- Rossella Filardi
- grid.4708.b0000 0004 1757 2822Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Giorgio Gargari
- grid.4708.b0000 0004 1757 2822Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Diego Mora
- grid.4708.b0000 0004 1757 2822Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Stefania Arioli
- grid.4708.b0000 0004 1757 2822Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| |
Collapse
|
39
|
Kumari M, Dasriya VL, Nataraj BH, Nagpal R, Behare PV. Lacticaseibacillus rhamnosus-Derived Exopolysaccharide Attenuates D-Galactose-Induced Oxidative Stress and Inflammatory Brain Injury and Modulates Gut Microbiota in a Mouse Model. Microorganisms 2022; 10:microorganisms10102046. [PMID: 36296322 PMCID: PMC9611687 DOI: 10.3390/microorganisms10102046] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 12/02/2022] Open
Abstract
This study aimed to investigate the protective effect of a novel exopolysaccharide EPSRam12, produced by Lacticaseibacillus rhamnosus Ram12, against D-galactose-induced brain injury and gut microbiota dysbiosis in mice. The findings demonstrate that EPSRam12 increases the level of antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase, total antioxidant capacity, and the level of anti-inflammatory cytokine IL-10, while decreasing malonaldehyde, nitric oxide, pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, MCP-1, and the mRNA expression of cyclooxygenase-2, inducible nitric oxide synthase, and the activation of nuclear factor-kappa-B in the brain tissues of D-galactose-treated mice. Further analyses reveal that EPSRam12 improves gut mucosal barrier function and increases the levels of short-chain fatty acids (SCFAs) in the intestine while restoring gut microbial diversity by enriching the abundance of SCFA-producing microbial genera Prevotella, Clostridium, Intestinimonas, and Acetatifactor while decreasing potential pathobionts including Helicobacter. These findings of antioxidative and anti-inflammatory effects in the brain and ameliorative effects on epithelial integrity, SCFAs and microbiota in the gut, provide novel insights into the effect of EPSRam12 intervention on the gut–microbiome–brain axis and should facilitate prospective understanding of microbial exopolysaccharide for improved host health.
Collapse
Affiliation(s)
- Manorama Kumari
- Technofunctional Starter Lab., National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, National Dairy Research Institute, Karnal 132001, Haryana, India
- College of Dairy and Food Technology, Agriculture University, Jodhpur 342304, Rajasthan, India
| | - Vaishali L. Dasriya
- Technofunctional Starter Lab., National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Basavaprabhu H. Nataraj
- Technofunctional Starter Lab., National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: (R.N.); (P.V.B.)
| | - Pradip V. Behare
- Technofunctional Starter Lab., National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, National Dairy Research Institute, Karnal 132001, Haryana, India
- Correspondence: (R.N.); (P.V.B.)
| |
Collapse
|
40
|
Singh S, Singh M, Gaur S. Probiotics as multifaceted oral vaccines against colon cancer: A review. Front Immunol 2022; 13:1002674. [PMID: 36263037 PMCID: PMC9573965 DOI: 10.3389/fimmu.2022.1002674] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics are known as the live microorganisms that, upon adequate administration, elicit a health beneficial response inside the host. The probiotics are known as immunomodulators and exhibit anti-tumor properties. Advanced research has explored the potential use of probiotics as the oral vaccines without the latent risks of pathogenicity. Probiotic-based oral vaccines are known to induce mucosal immunity that prevents the host from several enteric infections. Probiotic bacteria have the ability to produce metabolites in the form of anti-inflammatory cytokines, which play an important role in the prevention of carcinogenesis and in the activation of the phagocytes that eliminate the preliminary stage cancer cells. This review discusses the advantages and disadvantages of using the oral probiotic vaccines as well as the mechanism of action of probiotics in colon cancer therapy. This review also employs the use of “PROBIO” database for selecting certain probiotics with immunomodulatory properties. Furthermore, the use of several probiotic bacteria as anti-colon cancer adjuvants has also been discussed in detail. Because the current studies and trials are more focused on using the attenuated pathogens instead of using the probiotic-based vaccines, future studies must involve the advanced research in exploiting the potential of several probiotic strains as adjuvants in cancer therapies.
Collapse
Affiliation(s)
- Shubhi Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Manisha Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- *Correspondence: Smriti Gaur,
| |
Collapse
|
41
|
Detection of indigenous gut bacteria related to red chilli pepper (Capsicum annuum) in murine caecum and human faecal cultures. Mol Biol Rep 2022; 49:10239-10250. [PMID: 36068389 DOI: 10.1007/s11033-022-07875-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Red chilli pepper (Capsicum annuum; RP) is a popular spice containing the active compound capsaicin. Indigenous gut bacteria and metabolism can affect host health. The functions of capsaicin, including the regulation of metabolic health and anti-oxidant properties, may be correlated with the gut microbiota. METHODS To identify indigenous gut bacteria that are responsive to RP, Institute of Cancer Research mice fed a diet with no fibre or with 5% (w/w) RP for 14 days. Additionally, human stool samples collected from four healthy volunteers were incubated without (control) or with 2% (w/v) RP at 37 °C for 24 h. Microbiota in murine caecal samples and human faecal cultures were analysed using 16S rRNA (V4) amplicon sequencing. RESULTS Compared with the microbiota in mice fed no-fibre diets, Lachnospiraceae spp.-, Muribaculaceae spp.-, and Phacaeicola vulgatus-like bacteria were defined as murine RP-responsive indigenous gut bacteria (RP-RIB). In the human faecal cultures, acetate and propionate levels were higher in RP cultures than in the control cultures. Subdoligranulum spp.-, Blautia spp.-, Faecalibacterium prausnitzii-, P. vulgatus-, and Prevotella copri-like bacteria were defined as human RP-RIB. Compared with control culture Fe-reducing power was increased in the culture with RP. CONCLUSION RP increases the amount of short-chain fatty acid-producing bacteria and beneficial gut bacteria in mouse and human faecal cultures. Overall, RP could have a positive effect on the host by altering the gut microbiota.
Collapse
|
42
|
Kaur H, Kaur G, Ali SA. Dairy-Based Probiotic-Fermented Functional Foods: An Update on Their Health-Promoting Properties. FERMENTATION-BASEL 2022; 8:425. [DOI: 10.3390/fermentation8090425] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Numerous studies have shown a link between the consumption of fermented dairy foods and improved health outcomes. Since the early 2000s, especially probiotic-based fermented functional foods, have had a revival in popularity, mostly as a consequence of claims made about their health benefits. Among them, fermented dairy foods have been associated with obesity prevention and in other conditions such as chronic diarrhea, hypersensitivity, irritable bowel syndrome, Helicobacter pylori infection, lactose intolerance, and gastroenteritis which all are intimately linked with an unhealthy way of life. A malfunctioning inflammatory response may affect the intestinal epithelial barrier’s ability to function by interfering with the normal metabolic processes. In this regard, several studies have shown that fermented dairy probiotics products improve human health by stimulating the growth of good bacteria in the gut at the same time increasing the production of metabolic byproducts. The fermented functional food matrix around probiotic bacteria plays an important role in the survival of these strains by buffering and protecting them from intestinal conditions such as low pH, bile acids, and other harsh conditions. On average, cultured dairy products included higher concentrations of lactic acid bacteria, with some products having as much as 109/mL or g. The focus of this review is on fermented dairy foods and associated probiotic products and their mechanisms of action, including their impact on microbiota and regulation of the immune system. First, we discussed whey and whey-based fermented products, as well as the organisms associated with them. Followed by the role of probiotics, fermented-product-mediated modulation of dendritic cells, natural killer cells, neutrophils, cytokines, immunoglobulins, and reinforcement of gut barrier functions through tight junction. In turn, providing the ample evidence that supports their benefits for gastrointestinal health and related disorders.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, Indian Council of Agricultural Research-National Dairy Research Institute (ICAR-NDRI), Karnal 132001, India
| | - Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia
- Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
43
|
Apple Pomace Modulates the Microbiota and Increases the Propionate Ratio in an In Vitro Piglet Gastrointestinal Model. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8080408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Apple pomace (AP) contains biomolecules that induce changes in intestinal fermentation of monogastrics with positive expected health effects. The weaning of piglets can induce economic losses due to intestinal disturbances; new weaning strategies are, thus, welcome. The purpose of this study was to test the effect of AP on fermentation products by using baby-SPIME, an in vitro multi-compartment model dedicated to piglet weaning. A comparison was done on short chain fatty acid (SCFA) ratio and the microbiota induced in bioreactors between a control culture medium vs. an AP culture medium. The results of 2 preliminary runs showed that AP medium increased the molar ratio of propionate (p = 0.021) and decreased the molar ratio of butyrate (p = 0.009). Moreover, this medium increased the cumulative relative abundance of Prevotella sp. and Akkermansia sp. in bioreactors. AP could promote an ecosystem enriched with bacteria known as next-generation probiotics (NGP)—likely influencing the energy metabolism of piglets by their fermentation metabolites. AP could be used as a dietary strategy to influence bacterial changes in the intestine by stimulating the growth of bacteria identified as NGP.
Collapse
|
44
|
Gomez Quintero DF, Kok CR, Hutkins R. The Future of Synbiotics: Rational Formulation and Design. Front Microbiol 2022; 13:919725. [PMID: 35935226 PMCID: PMC9354465 DOI: 10.3389/fmicb.2022.919725] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Synbiotics, mixtures of live microbes and substrates selectively utilized by host organisms, are of considerable interest due to their ability to improve gastrointestinal health. However, formulating synbiotics remains challenging, due in part, to the absence of rational strategies to assess these products for synbiotic activities prior to clinical trials. Currently, synbiotics are formulated as either complementary or synergistic. Complementary synbiotics are made by combining probiotics and prebiotics, with each component acting independently and with the combination shown to provide a clinical health benefit. Most commercial synbiotics as well as those used in clinical trials have been of the complementary type. In contrast, synergistic synbiotics require that the added microbe is specifically stimulated or it’s persistence or activity are enhanced by the cognate substrate. Although several innovative examples have been described in the past few years based on this principle, in practice, relatively few synbiotic studies have tested for synergism. In this review, selected recent examples of complementary and synergistic synbiotics and the rationale for their formulation will be described. In addition, pre-clinical experimental approaches for identifying combinations that provide a basis for satisfying the requirements for synergism will be discussed.
Collapse
Affiliation(s)
- David F. Gomez Quintero
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Car Reen Kok
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, United States
- Complex Biosystems, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Robert Hutkins
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, United States
- *Correspondence: Robert Hutkins,
| |
Collapse
|
45
|
Lee SHF, Ahmad SR, Lim YC, Zulkipli IN. The Use of Probiotic Therapy in Metabolic and Neurological Diseases. Front Nutr 2022; 9:887019. [PMID: 35592636 PMCID: PMC9110960 DOI: 10.3389/fnut.2022.887019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/29/2022] [Indexed: 12/20/2022] Open
Abstract
The human gut is home to trillions of microbes that interact with host cells to influence and contribute to body functions. The number of scientific studies focusing on the gut microbiome has exponentially increased in recent years. Studies investigating factors that may potentially affect the gut microbiome and may be used for therapeutic purposes in diseases where dysbioses in the gut microbiome have been shown are of particular interest. This review compiles current evidence available in the scientific literature on the use of probiotics to treat metabolic diseases and autism spectrum disorders (ASDs) to analyze the efficacy of probiotics in these diseases. To do this, we must first define the healthy gut microbiome before looking at the interplay between the gut microbiome and diseases, and how probiotics affect this interaction. In metabolic diseases, such as obesity and diabetes, probiotic supplementation positively impacts pathological parameters. Conversely, the gut-brain axis significantly impacts neurodevelopmental disorders such as ASDs. However, manipulating the gut microbiome and disease symptoms using probiotics has less pronounced effects on neurodevelopmental diseases. This may be due to a more complex interplay between genetics and the environment in these diseases. In conclusion, the use of microbe-based probiotic therapy may potentially have beneficial effects in ameliorating the pathology of various diseases. Validation of available data for the development of personalized treatment regimens for affected patients is still required.
Collapse
Affiliation(s)
| | | | | | - Ihsan N. Zulkipli
- Pengiran Anak Puteri Rashidah Sa’adatul Bolkiah (PAPRSB) Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| |
Collapse
|
46
|
Manassi CF, de Souza SS, Hassemer GDS, Sartor S, Lima CMG, Miotto M, De Dea Lindner J, Rezzadori K, Pimentel TC, Ramos GLDPA, Esmerino E, Holanda Duarte MCK, Marsico ET, Verruck S. Functional meat products: Trends in pro-, pre-, syn-, para- and post-biotic use. Food Res Int 2022; 154:111035. [DOI: 10.1016/j.foodres.2022.111035] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
|
47
|
Dahiya D, Nigam PS. The Gut Microbiota Influenced by the Intake of Probiotics and Functional Foods with Prebiotics Can Sustain Wellness and Alleviate Certain Ailments like Gut-Inflammation and Colon-Cancer. Microorganisms 2022; 10:665. [PMID: 35336240 PMCID: PMC8954736 DOI: 10.3390/microorganisms10030665] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
The gut microbiota is composed of several microbial strains, with diverse and variable combinations in healthy and sick persons, changing at different stages of life. A healthy balance between host and gut microorganisms must be maintained in order to perform the normal physiological, metabolic, and immune functions and prevent disease development. Disturbances in the balance of the gut microbiota by diverse reasons initiate several health issues and promote the progression of certain diseases. This review is based on published research and reports that describe the role of probiotic microorganisms in the sustainability of health and the alleviation of certain diseases. Information is presented on the GRAS strains that are used as probiotics in the food industry for the production of fermented milk, yogurt, fermented food, functional foods, and probiotic drinks. To maintain a healthy microbiota, probiotic supplements in the form of freeze-dried live cells of probiotic strains are also available in different forms to consumers. The health benefits of lactic acid bacteria and other microorganisms and their role in the control of certain diseases such as gut inflammation, diabetes, and bowel cancer and in the safeguarding of the gut epithelial permeability from the invasion of pathogens are discussed.
Collapse
Affiliation(s)
- Divakar Dahiya
- Wexham Park Hospital, Wexham Street, Slough SL2 4HL, Berkshire, UK;
| | - Poonam Singh Nigam
- Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| |
Collapse
|
48
|
Zhao J, Wang L, Cheng S, Zhang Y, Yang M, Fang R, Li H, Man C, Jiang Y. A Potential Synbiotic Strategy for the Prevention of Type 2 Diabetes: Lactobacillus paracasei JY062 and Exopolysaccharide Isolated from Lactobacillus plantarum JY039. Nutrients 2022; 14:nu14020377. [PMID: 35057558 PMCID: PMC8782018 DOI: 10.3390/nu14020377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023] Open
Abstract
The disturbance of intestinal microorganisms and the exacerbation of type 2 diabetes (T2D) are mutually influenced. In this study, the effect of exopolysaccharides (EPS) from Lactobacillus plantarum JY039 on the adhesion of Lactobacillus paracasei JY062 was investigated, as well as their preventive efficacy against T2D. The results showed that the EPS isolated from L. plantarum JY039 effectively improved the adhesion rate of L. paracasei JY062 to Caco-2 cells (1.8 times) and promoted the proliferation of L. paracasei JY062. In the mice experiment, EPS, L. paracasei JY062 and their complex altered the structure of the intestinal microbiota, which elevated the proportion of Bifidobacterium, Faecalibaculum, while inversely decreasing the proportion of Firmicutes, Muribaculaceae, Lachnospiraceae and other bacteria involved in energy metabolism (p < 0.01; p < 0.05); enhanced the intestinal barrier function; promoted secretion of the gut hormone peptide YY (PYY) and glucagon-like peptide-1 (GLP-1); and reduced inflammation by balancing pro-inflammatory factors IL-6, TNF-α and anti-inflammatory factor IL-10 (p < 0.01; p < 0.05). These results illustrate that EPS and L. paracasei JY062 have the synbiotic potential to prevent and alleviate T2D.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chaoxin Man
- Correspondence: (C.M.); (Y.J.); Tel.: +86-18946196731(C.M.); +86-451-55191820(Y.J.)
| | - Yujun Jiang
- Correspondence: (C.M.); (Y.J.); Tel.: +86-18946196731(C.M.); +86-451-55191820(Y.J.)
| |
Collapse
|
49
|
Characterization of probiotic lactobacilli and development of fermented soymilk with improved technological properties. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2021.112827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
50
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|