1
|
Wang D, Xu R, Liu S, Sun X, Zhang T, Shi L, Wang Y. Enhancing the application of probiotics in probiotic food products from the perspective of improving stress resistance by regulating cell physiological function: A review. Food Res Int 2025; 199:115369. [PMID: 39658167 DOI: 10.1016/j.foodres.2024.115369] [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: 07/13/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Probiotic foods are foods containing probiotics, including dairy and non-dairy products, that exert significant beneficial impacts on human health. Benefiting from the rapid progress in systems biology, diverse types of probiotics with prominent health-promoting functionalities are unraveled, albeit such functions could be substantially influenced by the stress environments. Here, we conducted a comprehensive review to characterize the state-of-the-art research on probiotic foods and specific probiotics employed in their production. We summarized the detrimental effects of various environmental stresses, including those encountered during industrial fermentation and storage (in vitro), as well as in vivo conditions such as digestion and intestinal colonization, on the biological functions of probiotics. Furthermore, this review outlines the recent advancements in elucidating the mechanisms of stress resistance, which are expected to enhance targeted probiotic applications and optimize their functional properties. Additionally, we summarized various strategies aimed at improving stress tolerance by regulating cell physiological function, specifically adaptive laboratory evolution, preadaptation treatment, exogenous supplementation, and molecular biological manipulation. This review underscores the significance of enhancing our understanding of stress tolerance mechanisms at a systems level and developing efficacious anti-stress strategies to enhance the application of probiotics while maximizing their biological functionalities.
Collapse
Affiliation(s)
- Dingkang Wang
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ruijie Xu
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Sha Liu
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaomin Sun
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Tianxiao Zhang
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China.
| | - Youfa Wang
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
2
|
Liang W, Gao Y, Zhao Y, Gao L, Zhao Z, He Z, Li S. Lactiplantibacillus plantarum ELF051 Alleviates Antibiotic-Associated Diarrhea by Regulating Intestinal Inflammation and Gut Microbiota. Probiotics Antimicrob Proteins 2024; 16:1996-2006. [PMID: 37639209 PMCID: PMC11573863 DOI: 10.1007/s12602-023-10150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Probiotics are widely recognized for their ability to prevent and therapy antibiotic-associated diarrhea (AAD). This study was designed to evaluate Lactiplantibacillus plantarum ELF051 ability to prevent colon inflammation and its effect on gut microbial composition in a mouse model of AAD. The mice were intragastrically administered triple antibiotics for 7 days and then subjected to L. plantarum ELF051 for 14 days. The administration of L. plantarum ELF051 ameliorated the pathological changes in the colon tissue, downregulated interleukin (IL)-1β and tumor necrosis factor (TNF)-α, and upregulated IL-10, and increased the intestinal short-chain fatty acids (SCFAs) level. Lactiplantibacillus plantarum ELF051 also regulated the Toll-like receptor/myeloid differentiation primary response 88/nuclear factor kappa light chain enhancer of activated B cells (TLR4/MyD88/NF-κB) and the phosphatidylinositol 3-kinase/protein kinase B/ NF-κB (PI3K/AKT/ NF-κB) inflammatory signaling pathways. 16S rRNA analyses showed that L. plantarum ELF051 increased the abundance and diversity of gut bacteria, restoring gut microbiota imbalance. A Spearman's rank correlation analysis showed that lactobacilli are closely associated with inflammatory markers and SCFAs. This work demonstrated that L. plantarum ELF051 can attenuate antibiotic-induced intestinal inflammation in a mouse AAD model by suppressing the pro-inflammatory response and modulating the gut microbiota.
Collapse
Affiliation(s)
- Wei Liang
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yansong Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yujuan Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| | - Lei Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zijian Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zhongmei He
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
| | - Shengyu Li
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| |
Collapse
|
3
|
Liu S, Zhao S, Cheng Z, Ren Y, Shi X, Mu J, Ge X, Dai Y, Li L, Zhang Z. Akkermansia muciniphila Protects Against Antibiotic-Associated Diarrhea in Mice. Probiotics Antimicrob Proteins 2024; 16:1190-1204. [PMID: 37314693 DOI: 10.1007/s12602-023-10101-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 06/15/2023]
Abstract
Probiotics are used to prevent antibiotic-associated diarrhea (AAD) via the restoration of the gut microbiota. However, the precise effects of Akkermansia muciniphila (Akk), which is a promising probiotics, on AAD are unknown. Here, AAD models were established via the administration of lincomycin and ampicillin with or without pasteurized Akk or Amuc_1100 treatment. A diffusion test revealed that Akk was susceptible to the majority of the antibiotics, such as ampicillin. These effects were confirmed by the reduced Akk abundance in AAD model mice. Pasteurized Akk or Amuc_1100 significantly decreased the diarrhea status score and colon injury of AAD model mice. Additionally, these treatments significantly decreased the relative abundance of Citrobacter at genus level and reshaped the metabolic function of gut microbiota. Notably, pasteurized Akk or Amuc_1100 significantly changed the serum metabolome of AAD model mice. In addition, pasteurized Akk or Amuc_1100 suppressed intestinal inflammation by upregulating the expression of GPR109A and SLC5A8 and downregulating the expression of TNFα, IFNγ, IL1β, and IL6. Furthermore, they enhanced water and electrolyte absorption by upregulating AQP4, SLC26A3, and NHE3. Pasteurized Akk or Amuc_1100 also restored intestinal barrier function by ameliorating the downregulation of ZO-1, OCLN, CLDN4, and Muc2 in AAD model mice. In summary, optimizing intestinal health with pasteurized Akk or Amuc_1100 may serve as an approach for preventing AAD.
Collapse
Affiliation(s)
- Shenyin Liu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Suying Zhao
- Department of Laboratory Medicine, The Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, 210009, People's Republic of China
| | - Zhiwei Cheng
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yilin Ren
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xinyi Shi
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Jing Mu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xiangyang Ge
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Yuan Dai
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| | - Zhan Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
4
|
Gul S, Durante-Mangoni E. Unraveling the Puzzle: Health Benefits of Probiotics-A Comprehensive Review. J Clin Med 2024; 13:1436. [PMID: 38592298 PMCID: PMC10935031 DOI: 10.3390/jcm13051436] [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: 12/30/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 04/10/2024] Open
Abstract
A growing number of probiotic-containing products are on the market, and their use is increasing. Probiotics are thought to support the health of the gut microbiota, which in turn might prevent or delay the onset of gastrointestinal tract disorders. Obesity, type 2 diabetes, autism, osteoporosis, and some immunological illnesses are among the conditions that have been shown to possibly benefit from probiotics. In addition to their ability to favorably affect diseases, probiotics represent a defense system enhancing intestinal, nutritional, and oral health. Depending on the type of microbial strain utilized, probiotics can have variable beneficial properties. Although many microbial species are available, the most widely employed ones are lactic acid bacteria and bifidobacteria. The usefulness of these bacteria is dependent on both their origin and their capacity to promote health. Probiotics represent a valuable clinical tool supporting gastrointestinal health, immune system function, and metabolic balance. When used appropriately, probiotics may provide benefits such as a reduced risk of gastrointestinal disorders, enhanced immunity, and improved metabolic health. Most popular probiotics, their health advantages, and their mode of action are the topic of this narrative review article, aimed to provide the reader with a comprehensive reappraisal of this topic matter.
Collapse
Affiliation(s)
- Sabiha Gul
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via de Crecchio 7, 80138 Napoli, Italy;
| | - Emanuele Durante-Mangoni
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via de Crecchio 7, 80138 Napoli, Italy;
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli—Ospedale Monaldi, Piazzale Ettore Ruggieri, 80131 Napoli, Italy
| |
Collapse
|
5
|
Yan Z, Liu Z, Ma Y, Yang Z, Liu G, Fang J. Effects of Lactobacillus plantarum and Weissella viridescens on the Gut Microbiota and Serum Metabolites of Mice with Antibiotic-Associated Diarrhea. Nutrients 2023; 15:4603. [PMID: 37960257 PMCID: PMC10648191 DOI: 10.3390/nu15214603] [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: 09/04/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Antibiotic-associated diarrhea (AAD) refers to diarrhea caused by gut microbiota disorders after the use of antibiotics, which seriously threatens the health of humans and animals. Therefore, it is necessary to find an effective therapy to treat AAD. This research aimed to explore the effects of Lactobacillus plantarum H-6 (L. plantarum H-6) and Weissella viridescens J-1 (W. viridescens J-1) on alleviating antibiotic-associated diarrhea induced by lincomycin hydrochloride (LH) in mice. The results show that L. plantarum H-6 could significantly reduce the expression of pro-inflammatory factors such as IL-1β and IL-6 in colon tissue. At the same time, L. plantarum H-6 significantly increased the abundance of Lactobacillus and Akkermansia, decreased the abundance of Bacteroides, and increased the contents of L-tryptophan, LysoPC (20:4 (8Z, 11Z, 14Z, 17Z)), reduced riboflavin, threoninyl-methionine, and N-palmitoyl in serum. However, W. viridescens J-1 had little effect on the treatment of AAD. It can be concluded that L. plantarum H-6 can regulate mice's colonic microbial composition, improve their serum metabolic process, and alleviate antibiotic-associated diarrhea. This research may provide a novel therapeutic option for AAD.
Collapse
Affiliation(s)
- Zhiwei Yan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Zhuangzhuang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Yong Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Zhao Yang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; (Z.Y.); (Z.L.); (Y.M.); (Z.Y.); (G.L.)
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
6
|
Li W, Zhang S, Wang Y, Bian H, Yu S, Huang L, Ma W. Complex probiotics alleviate ampicillin-induced antibiotic-associated diarrhea in mice. Front Microbiol 2023; 14:1156058. [PMID: 37125182 PMCID: PMC10145528 DOI: 10.3389/fmicb.2023.1156058] [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: 02/01/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Aim Antibiotic-associated diarrhea (AAD) is a common side effect during antibiotic treatment, which can cause dysbacteriosis of the gut microbiota. Previous studies have shown beneficial effects in AAD treatment with Bifidobacterium lactis XLTG11, Lactobacillus casei Zhang, Lactobacillus plantarum CCFM8661, and Lactobacillus rhamnosus Probio-M9. However, no studies have been conducted on the immunomodulatory effects and protective intestinal barrier function of four complex probiotics. The aim of our study is to investigate the alleviation effects of complex probiotics on ampicillin-induced AAD. Methods Thirty-six BALB/c mice were randomly divided into six groups: normal control group (NC), model control group (MC), low-, medium-, and high-dose probiotics groups (LD, MD, and HD), and positive drug (Bifico, 1 × 107 cfu) control group (PDC; Bifico, also known as Bifidobacterium Triple Live Capsule, is composed of Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis). An AAD model was established by intragastric administration of ampicillin, by gavage of different doses of complex probiotics and Bifico. The weight gain, fecal water content, loose stool grade, intestinal permeability, total protein and albumin levels, intestinal barrier, cytokine levels, and gut microbiota were determined. Results The results showed that complex probiotics significantly decreased the fecal water content, loose stool grade, intestinal permeability, and ileum tissue damage. Their application increased the weight gain, SIgA, TP, and ALB levels. Additionally, complex probiotics significantly decreased the levels of pro-inflammatory cytokines and increased those of anti-inflammatory cytokines. Meanwhile, the mRNA expression levels of ZO-1, occludin, claudin-1, and MUC2 were significantly upregulated in the probiotic-treated group. Furthermore, the complex probiotics increased the gut microbiota diversity and modulated the changes in the gut microbiota composition caused by ampicillin. At the phylum level, the abundance of Proteobacteria in the HD group was lower than that in the MC group, whereas that of Bacteroidetes was higher. At the genus level, the abundances of Klebsiella and Parabacteroides in the HD group were lower, whereas those of Bacteroides, Muribaculaceae, and Lactobacillus were higher than those in the MC group. Moreover, Spearman's correlation analysis also found that several specific gut microbiota were significantly correlated with AAD-related indicators. Conclusion We found that complex probiotics improved the diarrhea-related indexes, regulated gut microbiota composition and diversity, increased the expression levels of intestinal protective barrier-related genes, preserved the intestinal barrier function, and relieved inflammation and intestinal injury, thereby effectively improving AAD-associated symptoms. Graphical Abstract.
Collapse
|
7
|
Huang YY, Wu JM, Wu WT, Lin JW, Liang YT, Hong ZZ, Jia XZ, Liu DM. Structural, antioxidant, and immunomodulatory activities of an acidic exopolysaccharide from Lactiplantibacillus plantarum DMDL 9010. Front Nutr 2022; 9:1073071. [PMID: 36570157 PMCID: PMC9779943 DOI: 10.3389/fnut.2022.1073071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
This study investigated the structural, antioxidant, and immunomodulatory activities of acidic exopolysaccharide (EPS-LP2) isolated from Lactiplantibacillus plantarum DMDL 9010. EPS-LP2 is composed of fucose (Fuc), arabinose (Ara), galactose (Gal), glucose (Glc), mannose (Man), and D-fructose (Fru) with a molar ratio of 0.13: 0.69: 8.32: 27.57: 62.07: 0.58: 0.46, respectively. Structural analysis of EPS-LP2 exhibited a smooth irregular lamellar surface, rod-like structure with swollen ends and slippery surfaces, and good thermal stability. Based on the methylation and NMR analysis, sugar residues including t-Manp, t-Glcp, 2-Manp, 6-Galp, 6-Glcp, and 4-Glcp were found to exist in EPS-LP2. In the 50∼400 μg/ml range, EPS-LP2 showed negligible neurotoxicity to RAW264.7 cells. Moreover, EPS-LP2 could protect RAW264.7 cells from oxidative injury by lowering the generation of reactive oxygen species (ROS), malondialdehyde (MDA), and the secretion of lactate dehydrogenase (LDH). In contrast, an increase in superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and the concentrations of glutathione (GSH) were observed. Immunoreactivity assays showed that EPS-LP2 could suppress the expression of NO, tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6) and inhibit the activation of the mitogen-activated protein kinase (MAPK)/nuclear factor-κB-gene binding (NF-κB) cell pathway. Conclusively, EPS-LP2 could be a potential natural antioxidant and immunomodulatory agent in functional foods and medicines.
Collapse
Affiliation(s)
- Yan-yan Huang
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Jia-min Wu
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Wei-tong Wu
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Jia-wei Lin
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Yan-tong Liang
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Zhen-zhen Hong
- College of Food Science and Engineering, Foshan University, Foshan, Guangdong, China,Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Xiang-ze Jia
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Dong-mei Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China,*Correspondence: Dong-mei Liu,
| |
Collapse
|
8
|
Kwon J, Kong Y, Wade M, Williams DJ, Creech CB, Evans S, Walter EB, Martin JM, Gerber JS, Newland JG, Hofto ME, Staat MA, Chambers HF, Fowler VG, Huskins WC, Pettigrew MM. Gastrointestinal Microbiome Disruption and Antibiotic-Associated Diarrhea in Children Receiving Antibiotic Therapy for Community-Acquired Pneumonia. J Infect Dis 2022; 226:1109-1119. [PMID: 35249113 PMCID: PMC9492313 DOI: 10.1093/infdis/jiac082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/02/2022] [Indexed: 11/14/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect of antibiotics. We examined the gastrointestinal microbiota in children treated with β-lactams for community-acquired pneumonia. Data were from 66 children (n = 198 samples), aged 6-71 months, enrolled in the SCOUT-CAP trial (NCT02891915). AAD was defined as ≥1 day of diarrhea. Stool samples were collected on study days 1, 6-10, and 19-25. Samples were analyzed using 16S ribosomal RNA gene sequencing to identify associations between patient characteristics, microbiota characteristics, and AAD (yes/no). Nineteen (29%) children developed AAD. Microbiota compositional profiles differed between AAD groups (permutational multivariate analysis of variance, P < .03) and across visits (P < .001). Children with higher baseline relative abundances of 2 Bacteroides species were less likely to experience AAD. Higher baseline abundance of Lachnospiraceae and amino acid biosynthesis pathways were associated with AAD. Children in the AAD group experienced prolonged dysbiosis (P < .05). Specific gastrointestinal microbiota profiles are associated with AAD in children.
Collapse
Affiliation(s)
- Jiye Kwon
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Yong Kong
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA.,Department of Molecular Biophysics and Biochemistry, W. M. Keck Foundation Biotechnology Resource Laboratory, Yale School of Medicine, New Haven, Connecticut, USA
| | - Martina Wade
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Derek J Williams
- Department of Pediatrics and the Vanderbilt Vaccine Research Program, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Clarence Buddy Creech
- Department of Pediatrics and the Vanderbilt Vaccine Research Program, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Scott Evans
- Biostatistics Center, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| | - Emmanuel B Walter
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Judy M Martin
- Department of Pediatrics, University of Pittsburgh School of Medicine and the UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey S Gerber
- Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason G Newland
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, Missouri, USA
| | - Meghan E Hofto
- Department of Pediatrics, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Mary Allen Staat
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Henry F Chambers
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Vance G Fowler
- Department of Medicine and Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - W Charles Huskins
- Mayo Clinic College of Medicine and Science and Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Melinda M Pettigrew
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | | |
Collapse
|
9
|
Bao W, He Y, Yu J, Liu M, Yang X, Ta N, Zhang E, Liang C. Regulatory Effect of Lactiplantibacillus plantarum 2-33 on Intestinal Microbiota of Mice With Antibiotic-Associated Diarrhea. Front Nutr 2022; 9:921875. [PMID: 35757257 PMCID: PMC9218693 DOI: 10.3389/fnut.2022.921875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Diarrhea is one of the common adverse reactions in antibiotic treatment, which is usually caused by the imbalance of intestinal flora, and probiotics play an important role in the structure of intestinal flora. Therefore, this experiment studied the regulatory effect of Lactiplantibacillus plantarum 2-33 on antibiotic-associated diarrhea (AAD) mice. First, the AAD mice model was established by the mixed antibiotic solution of gentamicin sulfate and cefradine. Then, the physiological indexes and diarrhea of mice were observed and recorded by gastric perfusion of low dose (1.0 × 107 CFU/ml), medium dose (1.0 × 108CFU/ml), and high dose (1.0 × 109 CFU/ml) strain 2-33. 16S rRNA gene V3-V4 regions were sequenced in colon contents of mice in control group, model group, self-healing group, and experimental group, respectively, and the diversity of intestinal flora and gene function prediction were analyzed. The results showed that the intestinal flora of AAD mice was not significantly regulated by gastric perfusion of strain 2-33 to 7 days, but the relative abundance and diversity of intestinal flora of AAD mice were significantly improved by gastric perfusion to 14 days (p < 0.05). In addition, at the genus level, the relative abundance of Lactobacillus increased significantly, and the relative abundance of Enterococcus and Bacillus decreased significantly (p < 0.05). In addition, the regulation of strain 2-33 on intestinal flora of AAD mice was time- and dose-dependent, short-term gastric perfusion, and low dose had no significant effect (p > 0.05). Strain 2-33 can significantly increase the levels of anti-inflammatory cytokines IL-4 and IL-10, significantly decrease the levels of proinflammatory cytokines TNF-α and IFN-γ (p < 0.05), and can also adjust carbohydrate metabolism, amino acid metabolism, and energy metabolism to normal levels, thus accelerating the recovery of intestinal flora structure of AAD mice. In summary, strain 2-33 can improve the structure and diversity of intestinal flora of AAD mice, balance the level of substance and energy metabolism, and play a positive role in relieving diarrhea, maintaining and improving the intestinal microecological balance.
Collapse
Affiliation(s)
- Wuyundalai Bao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuxing He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Jinghe Yu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Mingchao Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaofeng Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Na Ta
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Enxin Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Chengyuan Liang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
10
|
Xu B, Liang S, Zhao J, Li X, Guo J, Xin B, Li B, Huo G, Ma W. Bifidobacterium animalis subsp. lactis XLTG11 improves antibiotic-related diarrhea by alleviating inflammation, enhancing intestinal barrier function and regulating intestinal flora. Food Funct 2022; 13:6404-6418. [PMID: 35616024 DOI: 10.1039/d1fo04305f] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect during antibiotic treatment. In this study, we evaluated the regulatory effect of Bifidobacterium animalis subsp. lactis XLTG11 on mouse diarrhea caused by antibiotic-induced intestinal flora disturbance. Then, two strains of Bifidobacterium animalis subsp. lactis XLTG11 and Bifidobacterium animalis subsp. lactis BB-12 were administered to AAD mice. We found that the recovery effect of using B. lactis XLTG11 was better than that of B. lactis BB-12. B. lactis XLTG11 reduced the pathological characteristics of the intestinal tract, and significantly reduced the levels of lipopolysaccharide (LPS), D-lactic acid (D-LA) and diamine oxidase (DAO) to decrease intestinal permeability. In addition, these two strains significantly increased the expression of aquaporin and tight junction proteins, and inhibited toll-like receptor 4 (TLR4)/activation of the nuclear factor-κB (NF-κB) signaling pathway, significantly increased the levels of anti-inflammatory cytokines and decreased levels of pro-inflammatory cytokines. Moreover, after treatment with B. lactis XLTG11, the contents of acetic acid, propionic acid, butyric acid and total short-chain fatty acids were significantly increased. Compared with the MC group, B. lactis XLTG11 increased the abundance and diversity of the intestinal flora and changed the composition of the intestinal flora. We found that B. lactis XLTG11 can promote the recovery of intestinal flora and mucosal barrier function, thereby effectively improving AAD-related symptoms, providing a scientific basis for future clinical applications.
Collapse
Affiliation(s)
- Baofeng Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Shengnan Liang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayi Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Xuetong Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayao Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bowen Xin
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Haerbin 150000, China.
| |
Collapse
|
11
|
Roux E, Nicolas A, Valence F, Siekaniec G, Chuat V, Nicolas J, Le Loir Y, Guédon E. The genomic basis of the Streptococcus thermophilus health-promoting properties. BMC Genomics 2022; 23:210. [PMID: 35291951 PMCID: PMC8925076 DOI: 10.1186/s12864-022-08459-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/08/2022] [Indexed: 12/20/2022] Open
Abstract
Background Streptococcus thermophilus is a Gram-positive bacterium widely used as starter in the dairy industry as well as in many traditional fermented products. In addition to its technological importance, it has also gained interest in recent years as beneficial bacterium due to human health-promoting functionalities. The objective of this study was to inventory the main health-promoting properties of S. thermophilus and to study their intra-species diversity at the genomic and genetic level within a collection of representative strains. Results In this study various health-related functions were analyzed at the genome level from 79 genome sequences of strains isolated over a long time period from diverse products and different geographic locations. While some functions are widely conserved among isolates (e.g., degradation of lactose, folate production) suggesting their central physiological and ecological role for the species, others including the tagatose-6-phosphate pathway involved in the catabolism of galactose, and the production of bioactive peptides and gamma-aminobutyric acid are strain-specific. Most of these strain-specific health-promoting properties seems to have been acquired via horizontal gene transfer events. The genetic basis for the phenotypic diversity between strains for some health related traits have also been investigated. For instance, substitutions in the galK promoter region correlate with the ability of some strains to catabolize galactose via the Leloir pathway. Finally, the low occurrence in S. thermophilus genomes of genes coding for biogenic amine production and antibiotic resistance is also a contributing factor to its safety status. Conclusions The natural intra-species diversity of S. thermophilus, therefore, represents an interesting source for innovation in the field of fermented products enriched for healthy components that can be exploited to improve human health. A better knowledge of the health-promoting properties and their genomic and genetic diversity within the species may facilitate the selection and application of strains for specific biotechnological and human health-promoting purpose. Moreover, by pointing out that a substantial part of its functional potential still defies us, our work opens the way to uncover additional health-related functions through the intra-species diversity exploration of S. thermophilus by comparative genomics approaches. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08459-y.
Collapse
Affiliation(s)
- Emeline Roux
- INRAE, Institut Agro, STLO, Rennes, France.,Université de Lorraine, CALBINOTOX, Nancy, France.,Université de Rennes, INRIA, Campus de Beaulieu, Rennes, France
| | | | | | - Grégoire Siekaniec
- INRAE, Institut Agro, STLO, Rennes, France.,Université de Rennes, INRIA, Campus de Beaulieu, Rennes, France
| | | | - Jacques Nicolas
- Université de Rennes, INRIA, Campus de Beaulieu, Rennes, France
| | | | | |
Collapse
|
12
|
Huang YY, Wu YP, Jia XZ, Lin J, Xiao LF, Liu DM, Liang MH. Lactiplantibacillus plantarum DMDL 9010 alleviates dextran sodium sulfate (DSS)-induced colitis and behavioral disorders by facilitating microbiota-gut-brain axis balance. Food Funct 2022; 13:411-424. [PMID: 34913458 DOI: 10.1039/d1fo02938j] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous studies have found that probiotic supplements can ameliorate mental behavioral disorders. This study investigated the effects of Lactiplantibacillus plantarum DMDL 9010 (LP9010) intake on the depression-like behavior induced by dextran sodium sulfate (DSS) and its possible mechanism. Male C57BL/6N mice were fed with DSS to establish the model of ulcerative colitis. LP9010 intake reduced the DSS-induced inflammatory response, and repaired intestinal barrier damage, as well as lightened depression-like behavior. LP9010 supplementation also inhibited neuroinflammation by up-regulating the levels of neurotransmitters, especially 5-HT, NE, DA, and 5-HIAA. Moreover, the intake of LP9010 reorganized the gut microbiome by increasing the relative abundance of Bacteroidetes and Firmicutes, and decreasing the relative abundance of Proteobacteria and Verrucomicrobia. Furthermore, treatment with LP9010 increased the levels of short-chain fatty acids, such as butyric acid and propionic acid. In conclusion, LP9010 intake was a promising probiotic intervention strategy for the prevention of colitis-induced behavioral disorders through the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Yan-Yan Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| | - Ya-Ping Wu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| | - Xiang-Ze Jia
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China. .,Department of Health Sciences and Technology, ETH Zürich, Schmelzbergstrasse 9, Zürich, Switzerland
| | - Jin Lin
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| | - Lan-Fang Xiao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| | - Dong-Mei Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| | - Ming-Hua Liang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, Guangdong, China.
| |
Collapse
|
13
|
Xie J, Yuan Y, Tan H, Bai Y, Zheng Q, Mao L, Wu Y, Wang L, Da W, Ye Q, Zhang S, Wang J, Yin W, Bian Y, Ma W, Zhang L, Zhang R, Yu H, Guo Y. The combination of living Bifidobacterium, Lactobacillus, and Streptococcus improves social ranking and relieves anxiety-like behaviors in competitive mice in a social dominance tube test. Brain Behav 2022; 12:e2453. [PMID: 34878231 PMCID: PMC8785616 DOI: 10.1002/brb3.2453] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/03/2021] [Accepted: 11/06/2021] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Social rank has a profound influence on the behavior and health of humans and animals. METHODS To explore the effect of a combination of living Bifidobacterium, Lactobacillus and Streptococcus (CLB) on anxiety- and depression-like behaviors and social rank, mice were subjected to a social dominance tube test (SDTT). The behaviors, rank, gut microbiota, and expression of inflammatory cytokines and brain-derived neurotrophic factor (BDNF) in the hippocampus were measured. RESULTS The results indicated that CLB improved the SDTT ranking score of the losers and alleviated anxiety-like behaviors of the winners. CLB decreased the level of Desulfovibrio and augmented the level of Mollicutes in the feces, increased BDNF content, and reduced the level of tumor necrosis factor-α in the hippocampus. CONCLUSION These findings indicated that CLB may be used for the treatment of anxiety and improvement of the rank score via regulation of gut microbiota and anti-inflammatory effects.
Collapse
Affiliation(s)
- Jianping Xie
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China.,School of Pharmaceutical Science, Department of Zoology & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, P. R. China.,Library, Yunnan Minzu University, Kunming, Yunnan, P. R. China
| | - Yun Yuan
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Heng Tan
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Yufan Bai
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Qingyue Zheng
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Lin Mao
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Yushan Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Ling Wang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Wenhui Da
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Qingyan Ye
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Suting Zhang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Jing Wang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Wenyao Yin
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Yujing Bian
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Wenjie Ma
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Lanchun Zhang
- School of Pharmaceutical Science, Department of Zoology & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Rongping Zhang
- School of Pharmaceutical Science, Department of Zoology & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, P. R. China.,School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resources, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, P. R. China
| | - Haofei Yu
- School of Pharmaceutical Science, Department of Zoology & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Ying Guo
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, P. R. China
| |
Collapse
|
14
|
Desaka N, Ota C, Nishikawa H, Yasuda K, Ishii N, Bito T, Kishinaga Y, Naito Y, Higashimura Y. Streptococcus thermophilus extends lifespan through activation of DAF-16-mediated antioxidant pathway in Caenorhabditis elegans. J Clin Biochem Nutr 2022; 70:7-13. [PMID: 35068675 PMCID: PMC8764109 DOI: 10.3164/jcbn.21-56] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022] Open
Affiliation(s)
- Natsumi Desaka
- Department of Food Science, Ishikawa Prefectural University
| | - Chinatsu Ota
- United Graduate School of Agricultural Sciences, Tottori University
| | | | - Kayo Yasuda
- Department of Health Management, Tokai University Undergraduate School of Health Studies
| | - Naoaki Ishii
- Department of Health Management, Tokai University Undergraduate School of Health Studies
| | - Tomohiro Bito
- United Graduate School of Agricultural Sciences, Tottori University
| | - Yukio Kishinaga
- Research and Development Group, Mill Souhonsha Company Limited
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine
| | | |
Collapse
|