1
|
Polak K, Muszyński T, Frątczak A, Meznerics F, Bánvölgyi A, Kiss N, Miziołek B, Bergler-Czop B. Study of gut microbiome alterations in plaque psoriasis patients compared to healthy individuals. Postepy Dermatol Alergol 2024; 41:378-387. [PMID: 39290901 PMCID: PMC11404103 DOI: 10.5114/ada.2024.142394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/15/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Many studies have shown significant alterations in the gut microbiome of patients with psoriasis compared to healthy controls. Aim The primary objective of the current research was to explore the impact of gut microbiome composition on the progression and severity of plaque psoriasis. Material and methods A total of 20 patients with moderate-to-severe psoriasis and 20 healthy individuals were recruited and provided a stool sample to assess the gut microbiome. After the samples were prepared according to the NGS library preparation workflow, they were sequenced using the Illumina platform and the report was generated that underwent statistical analysis. Results The microbiome profiles of psoriasis patients exhibited significant differences compared to healthy controls as evidenced by the statistical analysis of various bacterial genera, with the median abundance significantly lower in psoriasis patients compared to healthy controls (p = 0.033). The analysis of the Firmicutes-to-Bacteroidetes ratio, a commonly evaluated marker of dysbiosis, did not reach statistical significance (p = 0.239). However, there was a noticeable trend towards a higher median ratio in psoriasis patients compared to healthy controls. The ratio did not show significant associations with PASI or BSA but trends towards significance with DLQI (B = -12.11, p = 0.095). Conclusions Overall, the above findings underscore the importance of the gut microbiome in psoriasis and suggest that modulation of specific bacterial genera, especially that with significant differences, could be a potential strategy for therapeutic intervention. Targeting these depleted genera through microbiome-based interventions, such as probiotic supplementation or faecal microbiota transplantation, could potentially help to restore gut homeostasis and alleviate the inflammatory burden in psoriasis.
Collapse
Affiliation(s)
- Karina Polak
- Department of Dermatology, Medical University of Silesia, Katowice, Poland
- Doctoral School of the Medical University of Silesia, Katowice, Poland
| | - Tomasz Muszyński
- Brothers Hospitallers of Saint John of God Hospital, Krakow, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University, Krakow, Poland
| | | | - Fanni Meznerics
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - András Bánvölgyi
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Norbert Kiss
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Bartosz Miziołek
- Department of Dermatology, Medical University of Silesia, Katowice, Poland
| | - Beata Bergler-Czop
- Department of Dermatology, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
2
|
Sanchez-Gallardo R, Bottacini F, Friess L, Esteban-Torres M, Somers C, Moore RL, McAuliffe FM, Cotter PD, van Sinderen D. Unveiling metabolic pathways of selected plant-derived glycans by Bifidobacterium pseudocatenulatum. Front Microbiol 2024; 15:1414471. [PMID: 39081887 PMCID: PMC11286577 DOI: 10.3389/fmicb.2024.1414471] [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: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Bifidobacteria are commonly encountered members of the human gut microbiota that possess the enzymatic machinery necessary for the metabolism of certain plant-derived, complex carbohydrates. In the current study we describe differential growth profiles elicited by a panel of 21 newly isolated Bifidobacterium pseudocatenulatum strains on various plant-derived glycans. Using a combination of gene-trait matching and comparative genome analysis, we identified two distinct xylanases responsible for the degradation of xylan. Furthermore, three distinct extracellular α-amylases were shown to be involved in starch degradation by certain strains of B. pseudocatenulatum. Biochemical characterization showed that all three α-amylases can cleave the related substrates amylose, amylopectin, maltodextrin, glycogen and starch. The genes encoding these enzymes are variably found in the species B. pseudocatenulatum, therefore constituting a strain-specific adaptation to the gut environment as these glycans constitute common plant-derived carbohydrates present in the human diet. Overall, our study provides insights into the metabolism of these common dietary carbohydrates by a human-derived bifidobacterial species.
Collapse
Affiliation(s)
- Rocio Sanchez-Gallardo
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Lisa Friess
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Maria Esteban-Torres
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Clarissa Somers
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Rebecca L. Moore
- UCD Perinatal Research Centre, School of Medicine, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, School of Medicine, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Paul D. Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Barcan AS, Barcan RA, Vamanu E. Therapeutic Potential of Fungal Polysaccharides in Gut Microbiota Regulation: Implications for Diabetes, Neurodegeneration, and Oncology. J Fungi (Basel) 2024; 10:394. [PMID: 38921380 PMCID: PMC11204944 DOI: 10.3390/jof10060394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/04/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
This review evaluates the therapeutic effects of polysaccharides derived from mushroom species that have medicinal and edible properties. The fungal polysaccharides were recently studied, focusing on their modulation of the gut microbiota and their impact on various diseases. The study covers both clinical and preclinical studies, detailing the results and highlighting the significant influence of these polysaccharides on gut microbiota modulation. It discusses the potential health benefits derived from incorporating these polysaccharides into the diet for managing chronic diseases such as diabetes, neurodegenerative disorders, and cancer. Furthermore, the review emphasizes the interaction between fungal polysaccharides and the gut microbiota, underscoring their role in modulating the gut microbial community. It presents a systematic analysis of the findings, demonstrating the substantial impact of fungal polysaccharides on gut microbiota composition and function, which may contribute to their therapeutic effects in various chronic conditions. We conclude that the modulation of the gut microbiota by these polysaccharides may play a crucial role in mediating their therapeutic effects, offering a promising avenue for further research and potential applications in disease prevention and treatment.
Collapse
Affiliation(s)
- Alexandru Stefan Barcan
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Graham Kerr Building, Glasgow G12 8QQ, UK
| | | | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
| |
Collapse
|
4
|
Samarra A, Cabrera-Rubio R, Martínez-Costa C, Collado MC. The role of Bifidobacterium genus in modulating the neonate microbiota: implications for antibiotic resistance acquisition in early life. Gut Microbes 2024; 16:2357176. [PMID: 38798019 PMCID: PMC11135851 DOI: 10.1080/19490976.2024.2357176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Resistance to antibiotics in newborns is a huge concern as their immune system is still developing, and infections and resistance acquisition in early life have short- and long-term consequences for their health. Bifidobacterium species are important commensals capable of dominating the infant gut microbiome and are known to be less prone to possess antimicrobial resistance genes than other taxa that may colonize infants. We aimed to study the association between Bifidobacterium-dominated infant gut microbiota and the antibiotic resistant gene load in neonates, and to ascertain the perinatal factors that may contribute to the antibiotic resistance acquisition. Two hundred infant fecal samples at 7 days and 1 month of age from the MAMI birth cohort were included in the study and for whom maternal-neonatal clinical records were available. Microbiota profiling was carried out by 16S rRNA amplicon sequencing, and targeted antibiotic resistance genes (ARGs) including tetM, tetW, tetO, blaTEM, blaSHV and ermB were quantified by qPCR. Infant microbiota clustered into two distinct groups according to their Bifidobacterium genus abundance: high and low. The main separation of groups or clusters at each time point was performed with an unsupervised non-linear algorithm of k-means partitioning to cluster data by time points based on Bifidobacterium genus relative abundance. Microbiota composition differed significantly between both groups, and specific bifidobacterial species were enriched in each cluster. Lower abundance of Bifidobacterium in the infant gut was associated with a higher load of antibiotic resistance genes. Our results highlight the relevance of Bifidobacterium genus in the early acquisition and establishment of antibiotic resistance in the gut. Further studies are needed to develop strategies to promote a healthy early colonization and fight against the spread of antibiotic resistances.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Raúl Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| |
Collapse
|
5
|
Jiménez-Arroyo C, Tamargo A, Molinero N, Reinosa JJ, Alcolea-Rodriguez V, Portela R, Bañares MA, Fernández JF, Moreno-Arribas MV. Simulated gastrointestinal digestion of polylactic acid (PLA) biodegradable microplastics and their interaction with the gut microbiota. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:166003. [PMID: 37549707 DOI: 10.1016/j.scitotenv.2023.166003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/06/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023]
Abstract
The accumulation of microplastics (MPs) in the environment as well as their presence in foods and humans highlight the urgent need for studies on the effects of these particles on humans. Polylactic acid (PLA) is the most widely used bioplastic in the food industry and medical field. Despite its biodegradability, biocompatibility, and "Generally Recognized As Safe" (GRAS) status, recent animal model studies have shown that PLA MPs can alter the intestinal microbiota; however, to date, no studies have been reported on the possible gut and health consequences of its intake by humans. This work simulates the ingestion of a realistic daily amount of PLA MPs and their pass through the gastrointestinal tract by combining the INFOGEST method and the gastrointestinal simgi® model to evaluate possible effects on the human colonic microbiota composition (16S rRNA gene sequencing analysis) and metabolic functionality (lactic acid and short-chain fatty acids (SCFA) production). Although PLA MPs did not clearly alter the microbial community homeostasis, increased Bifidobacterium levels tended to increase in presence of millimetric PLA particles. Furthermore, shifts detected at the functional level suggest an alteration of microbial metabolism, and a possible biotransformation of PLA by the human microbial colonic community. Raman spectroscopy and field emission scanning electron microscopy (FESEM) characterization revealed morphological changes on the PLA MPs after the gastric phase of the digestion, and the adhesion of organic matter as well as a microbial biofilm, with surface biodegradation, after the intestinal and colonic phases. With this evidence and the emerging use of bioplastics, understanding their impact on humans and potential biodegradation through gastrointestinal digestion and the human microbiota merits critical investigation.
Collapse
Affiliation(s)
- C Jiménez-Arroyo
- Institute of Food Science Research, CIAL, CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - A Tamargo
- Institute of Food Science Research, CIAL, CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - N Molinero
- Institute of Food Science Research, CIAL, CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - J J Reinosa
- Instituto de Cerámica y Vidrio, CSIC, c/ Kelsen, 28049 Madrid, Spain; Encapsulae S.L., c/Lituania 10, 12006 Castellón de la Plana, Spain
| | - V Alcolea-Rodriguez
- Instituto de Catálisis y Petroleoquímica, CSIC, c/ Marie Curie, 2, 28049 Madrid, Spain
| | - R Portela
- Instituto de Catálisis y Petroleoquímica, CSIC, c/ Marie Curie, 2, 28049 Madrid, Spain
| | - M A Bañares
- Instituto de Catálisis y Petroleoquímica, CSIC, c/ Marie Curie, 2, 28049 Madrid, Spain
| | - J F Fernández
- Encapsulae S.L., c/Lituania 10, 12006 Castellón de la Plana, Spain
| | - M V Moreno-Arribas
- Institute of Food Science Research, CIAL, CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain.
| |
Collapse
|
6
|
Chen JF, Ou-Yang MC, Hsia KC, Li CM, Yeh YT, Ho HH. A Three-Arm, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 Used Individually in Healthy Infants. Nutrients 2023; 15:3426. [PMID: 37571365 PMCID: PMC10421338 DOI: 10.3390/nu15153426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Probiotics are considered safe and beneficial to human health. However, the safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 in infants has not been confirmed. This study was to assess the safety of long-term oral administration of L. salivarius AP-32 and B. animalis CP-9 in healthy infants compared with placebo. A three-arm, randomized, double-blind, placebo-controlled trial was conducted in healthy, full-term infants. Eighty-eight infants between 7 days and 2 months (60 ± 7 days) of age were selected and randomized to treatment with L. salivarius AP-32, B. animalis CP-9 or placebo for 4 months. The unblinding indicated subjects were randomized to receive B. animalis CP-9 (N = 28), L. salivarius AP-32 (N = 29), or placebo (N = 31). A total of 76 infants completed the 4-month treatment with fully compliance. The primary outcome was weight gain, with no significant difference in infant weight at 4 months when comparing AP-32 or CP-9 group with the placebo group, either. The head circumference and recumbent length of the CP-9 group were not significantly different from those of the placebo group. The recumbent length of the AP-32 group was slightly lower than that in the placebo group at month 4, but there was no difference between the two groups in head circumference. Overall, the growth trend of all treatments was similar without significant difference. Furthermore, there were no apparent differences between each group in digestive tolerance, the occurrence of adverse events, crying/fussing time and episodes, alpha diversity, and beta diversity. The CP-9 group showed a significant increase in the abundance of the Bacteroides genus, while the AP-32 group demonstrated a significant increase in the abundance of the Lactobacillus genus when comparing the two probiotic groups. Our study findings indicate that the oral administration of both AP-32 and CP-9 strains has a positive impact on the maintenance of a healthy gut flora in infants. Long-term use of L. salivarius AP-32 or B. animalis CP-9 is safe for infants from 7 days to 6 months of age.
Collapse
Affiliation(s)
- Jui-Fen Chen
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Mei-Chen Ou-Yang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Ko-Chiang Hsia
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | | | - Ching-Min Li
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung 831, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung 831, Taiwan
| | - Hsieh-Hsun Ho
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| |
Collapse
|
7
|
Nishiyama K, Yong CC, Moritoki N, Kitazawa H, Odamaki T, Xiao JZ, Mukai T. Sharing of Moonlighting Proteins Mediates the Symbiotic Relationship among Intestinal Commensals. Appl Environ Microbiol 2023; 89:e0219022. [PMID: 36847513 PMCID: PMC10053696 DOI: 10.1128/aem.02190-22] [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: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of symbiotic bacteria that form a complex ecological community and influence human physiology. Symbiotic nutrient sharing and nutrient competition are the most studied relationships in gut commensals, whereas the interactions underlying homeostasis and community maintenance are not fully understood. Here, we provide insights into a new symbiotic relationship wherein the sharing of secreted cytoplasmic proteins, called "moonlighting proteins," between two heterologous bacterial strains (Bifidobacterium longum and Bacteroides thetaiotaomicron) was observed to affect the adhesion of bacteria to mucins. B. longum and B. thetaiotaomicron were cocultured using a membrane-filter system, and in this system the cocultured B. thetaiotaomicron cells showed greater adhesion to mucins compared to that shown by monoculture cells. Proteomic analysis showed the presence of 13 B. longum-derived cytoplasmic proteins on the surface of B. thetaiotaomicron. Moreover, incubation of B. thetaiotaomicron with the recombinant proteins GroEL and elongation factor Tu (EF-Tu)-two well-known mucin-adhesive moonlighting proteins of B. longum-led to an increase in the adhesion of B. thetaiotaomicron to mucins, a result attributed to the localization of these proteins on the B. thetaiotaomicron cell surface. Furthermore, the recombinant EF-Tu and GroEL proteins were observed to bind to the cell surface of several other bacterial species; however, the binding was species dependent. The present findings indicate a symbiotic relationship mediated by the sharing of moonlighting proteins among specific strains of B. longum and B. thetaiotaomicron. IMPORTANCE The adhesion of intestinal bacteria to the mucus layer is an important colonization strategy in the gut environment. Generally, the bacterial adhesion process is a characteristic feature of the individual cell surface-associated adhesion factors secreted by a particular bacterium. In this study, coculture experiments between Bifidobacterium and Bacteroides show that the secreted moonlighting proteins adhere to the cell surface of coexisting bacteria and alter the adhesiveness of the bacteria to mucins. This finding indicates that the moonlighting proteins act as adhesion factors for not only homologous strains but also for coexisting heterologous strains. The presence of a coexisting bacterium in the environment can significantly alter the mucin-adhesive properties of another bacterium. The findings from this study contribute to a better understanding of the colonization properties of gut bacteria through the discovery of a new symbiotic relationship between them.
Collapse
Affiliation(s)
- Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Cheng-Chung Yong
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
8
|
Hammouda ZK, Wasfi R, Abdeltawab NF. Hormonal drugs: Influence on growth, biofilm formation, and adherence of selected gut microbiota. Front Cell Infect Microbiol 2023; 13:1147585. [PMID: 36992682 PMCID: PMC10042233 DOI: 10.3389/fcimb.2023.1147585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/17/2023] [Indexed: 03/14/2023] Open
Abstract
Many studies have reported the influence of hormonal drugs on gut microbiota composition. However, the underlying mechanism of this interaction is still under study. Therefore, this study aimed to evaluate the possible in vitro changes in selected members of gut bacteria exposed to oral hormonal drugs used for years. Selected members of gut bacteria were Bifidobacterium longum, Limosilactobacillus reuteri, Bacteroides fragilis, and Escherichia coli representing the four main phyla in the gut. Selected hormonal drugs used for a long time were estradiol, progesterone, and thyroxine. The effect of intestinal concentrations of these drugs on the selected bacterial growth, biofilm formation, and adherence to Caco-2/HT-29 cell line was assessed. Short-chain fatty acids (SCFAs) have been included in host functions including the gut, immune and nervous functions; thus, the drug’s effects on their production were assayed using High- Performance Liquid Chromatography. Sex steroids significantly increased the growth of all tested bacteria except B. longum, similarly, thyroxine increased the growth of tested Gram-negative bacteria however reducing that of tested Gram-positive bacteria. The effect of drugs on biofilm formation and bacterial adherence to cell lines cocultures was variable. Progesterone decreased the biofilm formation of tested Gram-positive bacteria, it nevertheless increased L. reuteri adherence to Caco-2/HT-29 cell line cell lines coculture. By contrast, progesterone increased biofilm formation by Gram-negative bacteria and increased adherence of B. fragilis to the cell lines coculture. Moreover, thyroxine and estradiol exhibited antibiofilm activity against L. reuteri, while thyroxine increased the ability of E. coli to form a biofilm. Moreover, hormones affected bacterial adherence to cell lines independently of their effect on hydrophobicity suggesting other specific binding factors might contribute to this effect. Tested drugs affected SCFAs production variably, mostly independent of their effect on bacterial growth. In conclusion, our results showed that the microbiota signature associated with some hormonal drug consumption could be the result of the direct effect of these drugs on bacterial growth, and adherence to enterocytes besides the effect of these drugs on the host tissue targets. Additionally, these drugs affect the production of SCFAs which could contribute to some of the side effects of these drugs.
Collapse
Affiliation(s)
- Zainab K. Hammouda
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Reham Wasfi
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
- *Correspondence: Reham Wasfi,
| | - Nourtan F. Abdeltawab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Development of the Anaerobic Microbiome in the Infant Gut. Pediatr Infect Dis J 2023:00006454-990000000-00384. [PMID: 36917032 DOI: 10.1097/inf.0000000000003905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Ninety-five percent of gut microbiota are anaerobes and vary according to age and diet. Complex carbohydrates in human milk enhance the growth of Bifidobacterium and Bacteroides in the first year. Complex carbohydrates in solid foods enhance the growth of Bacteroides and Clostridium in the second year. Short-chain fatty acids produced by Akkermansia and Faecalibacterium may reduce obesity, diabetes and IBD.
Collapse
|
10
|
Tardiolo G, Romeo O, Zumbo A, Di Marsico M, Sutera AM, Cigliano RA, Paytuví A, D’Alessandro E. Characterization of the Nero Siciliano Pig Fecal Microbiota after a Liquid Whey-Supplemented Diet. Animals (Basel) 2023; 13:642. [PMID: 36830429 PMCID: PMC9951753 DOI: 10.3390/ani13040642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
The utilization of dairy by-products as animal feed, especially in swine production, is a strategy to provide functional ingredients to improve gut health. This study explored the potential effect of a liquid whey-supplemented diet on the fecal microbiota of eleven pigs belonging to the Nero Siciliano breed. Five pigs were assigned to the control group and fed with a standard formulation feed, whereas six pigs were assigned to the experimental group and fed with the same feed supplemented with liquid whey. Fecal samples were collected from each individual before the experimental diet (T0), and one (T1) and two (T2) months after the beginning of the co-feed supplementation. Taxonomic analysis, based on the V3-V4 region of the bacterial 16S rRNA, showed that pig feces were populated by a complex microbial community with a remarkable abundance of Firmicutes, Bacteroidetes, and Spirochaetes phyla and Prevotella, Lactobacillus, Clostridium, and Treponema genera. Alpha and beta diversity values suggested that the experimental diet did not significantly affect the overall fecal microbiota diversity. However, analysis of abundance at different time points revealed significant variation in several bacterial genera, suggesting that the experimental diet potentially affected some genera of the microbial community.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | - Orazio Romeo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Alessandro Zumbo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | - Marco Di Marsico
- Sequentia Biotech SL, Carrer del Dr. Trueta 179, 08005 Barcelona, Spain
| | - Anna Maria Sutera
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | | | - Andreu Paytuví
- Sequentia Biotech SL, Carrer del Dr. Trueta 179, 08005 Barcelona, Spain
| | - Enrico D’Alessandro
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| |
Collapse
|
11
|
Wang HY, Chen S, Xue RY, Lin XY, Yang JL, Zhang YS, Li SW, Juhasz AL, Ma LQ, Zhou D, Li HB. Arsenic Ingested Early in Life Is More Readily Absorbed: Mechanistic Insights from Gut Microbiota, Gut Metabolites, and Intestinal Morphology and Functions. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:1017-1027. [PMID: 36580282 DOI: 10.1021/acs.est.2c04584] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Early-life arsenic (As) exposure is a particular health concern. However, it is unknown if As ingested early in life is more readily absorbed from the gastrointestinal (GI) tract, i.e., higher in oral bioavailability. Here, weanling (3-week) and adult (6-week-old) female mice were exposed to arsenate in the diet (10 μg g-1) over a 3-week period with As oral bioavailability estimated using As urinary excretion as the bioavailability endpoint. The As urinary excretion factor was 1.54-fold higher in weanling mice compared to adult mice (82.2 ± 7.29 versus 53.1 ± 3.73%), while weanling mice also showed 2.28-, 1.50-, 1.48-, and 1.89-fold higher As concentration in small intestine tissue, blood, liver, and kidneys, demonstrating significantly higher As oral bioavailability of early-life exposure. Compared to adult mice, weanling mice significantly differed in gut microbiota, but the difference did not lead to remarkable differences in As biotransformation in the GI tract or tissue and in overall gut metabolite composition. Although the expression of several metabolites (e.g., atrolactic acid, hydroxyphenyllactic acid, and xanthine) was up-regulated in weanling mice, they had limited ability to elevate As solubility in the intestinal tract. Compared to adult mice, the intestinal barrier function and intestinal expression of phosphate transporters responsible for arsenate absorption were similar in weanling mice. However, the small intestine of weanling mice was characterized by more defined intestinal villi with greater length and smaller width, providing a greater surface area for As to be absorbed across the GI barrier. The results highlight that early-life As exposure can be more readily absorbed, advancing the understanding of its health risk.
Collapse
Affiliation(s)
- Hong-Yu Wang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Shan Chen
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Rong-Yue Xue
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xin-Ying Lin
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Jin-Lei Yang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yao-Sheng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Shi-Wei Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Albert L Juhasz
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Lena Q Ma
- Institute of Soil and Water Resources and Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hong-Bo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|
12
|
Banavar A, Amirkolaei SK, Duscher L, Khairunisa BH, Mukhopadhyay B, Schwarz M, Urick S, Ovissipour R. Nutritional Evaluation of Black Soldier Fly Frass as an Ingredient in Florida Pompano (Trachinotus carolinus L.) Diets. Animals (Basel) 2022; 12:ani12182407. [PMID: 36139267 PMCID: PMC9495079 DOI: 10.3390/ani12182407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/15/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Carnivorous fish species, such as the Florida pompano, currently require expensive and unsustainable feed in order to be raised in an aquaculture setting. A waste product of the black soldier fly, frass has the potential to be an alternative carbohydrate source for these fish. This study aims to assess the potential of frass to mimic the growth, body, composition, and intestinal microbiome of pompano that can be achieved through commercial feed. Three diets with varying frass levels as well as a control diet were developed. The frass was found to increase the visceral somatic index and lower the hepatosomatic index. The frass did not alter the body composition or improve the growth performance, leading to a lower specific growth rate and higher feed conversion rate. The microbiome analysis showed the highest diversity of the gut flora in the control diet, while the frass diets showed signs of community imbalance. This may have been due to the decreased starch in the frass, which is usually metabolized by those microbial communities. Overall, frass diets are not ideal for carnivorous fish diets, but have could have potential for feed replacement in herbivore and detritivore fish species. Abstract The aquaculture industry is in need of sustainable fish feed to reduce the use of expensive and environmentally invasive wild-caught fish currently fed to many carnivorous species. The black soldier fly (BSF) has become a popular sustainable alternative protein source; however, the nutritional waste byproduct of BSF, frass, has not been extensively studied as a feed replacement in carnivorous species. This study evaluates the potential of BSF frass on the growth, body composition, and intestinal microbiome of the Florida pompano, Trachinotus carolinus. Four experimental diets were formulated containing different levels of frass, replacing plant-based carbohydrate sources. As a result of this study, the frass did not improve the growth performance, resulting in a lower specific growth rate and higher feed conversion rate. While the frass diets did not alter the body composition, the visceral somatic index (VSI) significantly increased compared to the control diet and the hepatosomatic index (HIS) was lowered. The microbiome analysis showed high variation among the diets, with the control diet having the most distinct consortia, which may have been driven by the increased levels of starch compared to frass diets. This study indicates that BSF frass may not be a suitable feed replacement for carnivorous pompano; however, frass could still potentially be a replacement feed for herbivore or detritivore fish and should be further studied.
Collapse
Affiliation(s)
- Amiti Banavar
- Future Foods Lab and Cellular Agriculture Initiative, Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Samad Keramat Amirkolaei
- Future Foods Lab and Cellular Agriculture Initiative, Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
| | - Lexi Duscher
- Future Foods Lab and Cellular Agriculture Initiative, Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
| | - Bela Haifa Khairunisa
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Biswarup Mukhopadhyay
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Michael Schwarz
- Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
| | - Steve Urick
- Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
| | - Reza Ovissipour
- Future Foods Lab and Cellular Agriculture Initiative, Virginia Seafood Agricultural Research and Extension Center, Virginia Polytechnic Institute and State University, Hampton, VA 23699, USA
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Biological Systems Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Correspondence:
| |
Collapse
|
13
|
Combination of microbiome and metabolome to analyze the cross-synergism mechanism of inulin and gut microbiota in vitro and vivo. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
14
|
Chang SC, Chiang HH, Liu CY, Li YJ, Lu CL, Lee YP, Huang CJ, Lai CL. Intestinal Mucosal Barrier Improvement with Prebiotics: Histological Evaluation of Longish Glucomannan Hydrolysates-Induced Innate T Lymphocyte Activities in Mice. Nutrients 2022; 14:nu14112220. [PMID: 35684019 PMCID: PMC9182621 DOI: 10.3390/nu14112220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Use of prebiotics is a growing topic in healthcare. A lightweight molecule and water-soluble fiber ingredient, longish glucomannan hydrolysates (LGH), has been developed to improve the intestinal mucosal barrier and confer gut health benefits. This study aims to investigate the implications of continuous LGH intervening in intestinal epithelium integrity and protective immunity against chemical dextran sodium sulfate (DSS)-induced colitis. Twelve male BALB/c mice were randomly arranged into four groups. The LGH/DSS group had results in bodyweight variance, epithelial cell density, and aberrancy score as good as the LGH group, and both were equivalent to the control group. LGH consumption effectively protects the distal intestinal epithelium by activating innate T lymphocytes. Meanwhile, T-cell subsets in subepithelial interspersion take a bystander role in these microenvironmental alterations. Under this stress, the cluster of differentiation 3 (CD3)+ T cells infiltrate the epithelium, while CD4+ T cells inversely appear in submucosal large lymphoid aggregates/isolated lymphoid follicles (ILFs) in which significant CD3+, CD4+, and CD8+ T-cell populations agglomerate. Moreover, forkhead box P3 (Foxp3) and interleukin 17 (IL-17) are observed in these ILFs. Agglomerated CD4+ T-cell lineages may have roles with proinflammatory T helper 17 cells and anti-inflammatory regulatory T cells in balancing responses to intraluminal antigens. Collectively, LGH administration may function in immune modulation to protect against DSS-induced inflammation.
Collapse
Affiliation(s)
- Shih-Chang Chang
- Division of Colorectal Surgery, Department of Surgery, Cathay General Hospital, Taipei 106438, Taiwan;
| | - Hui-Hsun Chiang
- School of Nursing, National Defense Medical Center, Taipei 114201, Taiwan;
| | - Chih-Yi Liu
- Division of Pathology, Sijhih Cathay General Hospital, New Taipei City 221037, Taiwan;
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Yu-Ju Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Chung-Lun Lu
- Aquatic Technology Research Center, Agricultural Technology Research Institute, Xiangshan, Hsinchu 300110, Taiwan;
| | - Yung-Pin Lee
- Research and Development, Healthy-Bioceuticals Company, Taipei 114201, Taiwan;
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Taipei 106438, Taiwan;
- Department of Biochemistry, National Defense Medical Center, Taipei 114201, Taiwan
| | - Ching-Long Lai
- Division of Basic Medical Sciences, Department of Nursing, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
- Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Tarracchini C, Viglioli M, Lugli GA, Mancabelli L, Fontana F, Alessandri G, Turroni F, Ventura M, Milani C. The Integrated Probiotic Database: a genomic compendium of bifidobacterial health-promoting strains. MICROBIOME RESEARCH REPORTS 2022; 1:9. [PMID: 38045645 PMCID: PMC10688828 DOI: 10.20517/mrr.2021.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 02/07/2022] [Indexed: 12/05/2023]
Abstract
Background: The World Health Organization defines probiotics as "live microorganisms, which when administered in adequate amounts confer a health benefit on the host". In this framework, probiotic strains should be regarded as safe for human and animal consumption, i.e., they should possess the GRAS (generally recognized as safe) status, notified by the local authorities. Consistently, strains of selected Bifidobacterium species are extensively used as probiotic agents to prevent and ameliorate a broad spectrum of human and/or animal gastrointestinal disorders. Even though probiotic properties are often genus- or species-associated, strain-level differences in the genetic features conferring individual probiotic properties to commercialized bifidobacterial strains have not been investigated in detail. Methods: In this study, we built a genomic database named Integrated Probiotic DataBase (IPDB), whose first iteration consists of common bifidobacterial strains used in probiotic products for which public genome sequences were available, such as members of B. longum subsp. longum, B. longum subsp. infantis, B. bifidum, B. breve, and B. animalis subsp. lactis taxa. Furthermore, the IPDB was exploited to perform comparative genome analyses focused on genetic factors conferring structural, functional, and chemical features predicted to be involved in microbe-host and microbe-microbe interactions. Results and conclusion: Our analyses revealed strain-level genetic differences, underlining the importance of inspecting the strain-specific and outcome-specific efficacy of probiotics. In this context, IPDB represents a valuable resource for obtaining genetic information of well-established bifidobacterial probiotic strains.
Collapse
Affiliation(s)
- Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Martina Viglioli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
- GenProbio Srl, Via delle Scienze, 11/A, Parma 43100, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11a, Parma 43124, Italy
| |
Collapse
|
16
|
Immunomodulatory Activity of Extracellular Vesicles of Kimchi-Derived Lactic Acid Bacteria ( Leuconostoc mesenteroides, Latilactobacillus curvatus, and Lactiplantibacillus plantarum). Foods 2022; 11:foods11030313. [PMID: 35159463 PMCID: PMC8834128 DOI: 10.3390/foods11030313] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 12/12/2022] Open
Abstract
Lactic acid bacteria present in Kimchi, such as Leuconostoc mesenteroides (Lm), Latilactobacillus curvatus (Lc), and Lactiplantibacillus plantarum (Lp) produce extracellular vesicles (ECVs) that modulate immune responses. The ECVs of probiotic Kimchi bacteria are abbreviated as LmV, LcV, and LpV. Treatment of macrophages (RAW264.7) with ECVs (LmV, LcV, and LpV) increased the production of nitric oxide (NO), tumor necrosis factor (TNF)-α, and interleukin-6 (IL-6). Immunostimulatory effects exerted on the RAW264.7 cells were stronger after treatments with LmV and LcV than with LpV. Treatment of mice with LcV (1 mg/kg, orally) induced splenocyte proliferation and subsequent production of both NO and cytokines (INF-γ, TNF-α, IL-4, and IL-10). Furthermore, pre-treatment of macrophages and microglial cells with ECVs prior to LPS stimulation significantly attenuated the production of NO and pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). Therefore, ECVs (LmV, LcV, and LpV) prevent inflammatory responses in the LPS-stimulated microglial cells by blocking the extracellular signal-regulated kinase (Erk) and p38 signaling pathways. These results showed that LmV, LcV, and LpV from Kimchi probiotic bacteria safely exert immunomodulatory effects.
Collapse
|
17
|
Liu C, Jiang W, Yang F, Guo Y, Yao W, Cheng Y, Zhao Y, He Q. Combination of microbiome and metabolome to analyze the cross-cooperation mechanism of Echinacea purpurea polysaccharide with gut microbiota in vitro and in vivo. Food Funct 2022; 13:10069-10082. [DOI: 10.1039/d2fo02336a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Echinacea purpurea polysaccharide (EPP) is a functional compound in Echinacea purpurea. At the present, it is generally recognized that plant polysaccharides can regulate the intestinal microecology, but there are few...
Collapse
|