1
|
Ismael M, Qayyum N, Gu Y, Na L, Haoyue H, Farooq M, Wang P, Zhong Q, Lü X. Functional Effects of Probiotic Lactiplantibacillus plantarum in Alleviation Multidrug-Resistant Escherichia coli-Associated Colitis in BALB/c Mice Model. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10356-7. [PMID: 39271561 DOI: 10.1007/s12602-024-10356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
Multidrug-resistant Escherichia coli (MDR-E. coli) is a global health concern. Lactic acid bacteria (LAB) are important probiotics that have beneficial effects on health, and in recent years, their influences in preventing foodborne pathogens-induced colitis have attracted much attention. Therefore, this study aimed to investigate the oral administration of Lactiplantibacillus plantarum NWAFU-BIO-BS29 as an emerging approach to alleviate MDR-E. coli-induced colitis in BALB/c mice model. To illustrate the mode of action of NWAFU-BIO-BS29 interventions with the gut microbiota and immune responses, the changes on the colonic mucosal barrier, regulatory of the gene expressions of inflammatory cytokines, re-modulating the intestinal microflora, and changes in physiological parameters were studied. The results indicated that daily supplementation of 200 µL fresh bacteria for 7 days had ameliorated the associated colitis and partially prevented the infection. The modes of action by ameliorating the inflammatory response, which destructed villous and then affected the intestinal barrier integrity, reducing the secretion of interleukins (6 and β) and tumor necrosis factor (TNF-α) in serum by 87.88-89.93%, 30.73-35.98%, and 19.14-22.32%, respectively, enhancing the expressions of some epithelial integrity-related proteins in the mouse mucous layer of mucins 2 and 3, Claudin-1, and Occludin by 130.00-661.85%, 27.64-57.35%, 75.52-162.51%, and 139.36-177.73%, respectively, and 56.09-73.58% for toll-like receptor (TLR4) in colon tissues. Notably, the mouse gut microbiota analysis showed an increase in the relative abundance of beneficial bacteria, including Lactobacillus, Bacteriodales bacterium, Candidatus Saccharimonas, Enterorhabdus, and Bacilli. Furthermore, the probiotic promoted the proliferation of epithelia and goblet cells by increasing short-chain fatty acids (SCFAs) levels by 19.23-31.39%. In conclusion, L. plantarum NWAFU-BIO-BS29 has potential applications and can be considered a safe dietary supplement to ameliorate the colitis inflammation symptoms of MDR-E. coli infection.
Collapse
Affiliation(s)
- Mohamedelfatieh Ismael
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
- Sudanese Standards and Metrology Organization, Khartoum, 13573, Sudan
| | - Nageena Qayyum
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Yaxin Gu
- College of Food Science, China Agricultural University, Beijing, China
| | - Li Na
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Han Haoyue
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Muhammad Farooq
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Panpan Wang
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Qingping Zhong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Xin Lü
- Lab of Bioresource, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
2
|
Altves S, Celik FS, Gunes CE, Bilecen K. Exploring the therapeutic potential of Leuconostoc mesenteroides lysates in wound healing and immune modulation on keratinocyte cells. Arch Dermatol Res 2024; 316:548. [PMID: 39162738 DOI: 10.1007/s00403-024-03324-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 06/26/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The skin, being the body's largest organ, primarily functions as a formidable defense mechanism against potential microbial infections. The skin's microbiota, consisting of a complex assembly of microorganisms, exerts a pivotal influence on skin homeostasis by modulating keratinocytes and their cytokine secretion, thereby playing an integral role in promoting optimal cutaneous health. Leuconostoc mesenteroides finds extensive application in the production of fermented foods and bacteriocins. Empirical studies validate the effectiveness of L. mesenteroides treatments in enhancing immune function and demonstrating notable antioxidant characteristics. This study investigates the potential of L. mesenteroides in improving skin health and wound healing. It also aims to comprehend their impact on wound healing markers, cytokine production, and cell cycle regulation compared to ferulic acid, known for its wound healing effects. Our findings indicate that L. mesenteroides lysate possesses antibacterial properties against Staphylococcus aureus and Pseudomonas aeruginosa, along with the ability to mitigate their toxic effects in a pathogen-simulating model employing HaCaT keratinocyte cells. Additionally, the lysate demonstrated noteworthy wound closure after a 24-hour treatment, along with a significant reduction in interleukin-6 levels and oxidative stress index. Modulation of the cell cycle is evident by decreasing G0/G1 phases and increasing S and G2/M phases and enhanced expression of wound healing marker genes and proteins CDH1. In conclusion, L. mesenteroides lysate exhibits immune-modulating and antibacterial properties, offering potential alternatives to conventional treatments for various skin conditions. These findings contribute to the exploration of innovative approaches to enhancing human life through skin health and wound healing.
Collapse
Affiliation(s)
- Safaa Altves
- Science and Technology Research and Application Center (BITAM), Necmettin Erbakan University, Konya, Türkiye.
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye.
| | - Fatma Secer Celik
- Department of Medical Biology, Faculty of Medicine, Ankara Medipol University, Ankara, Türkiye
| | - Canan Eroglu Gunes
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye
| | - Kivanc Bilecen
- Department of Molecular Biology and Genetics, Faculty of Agriculture and Natural Sciences, Konya Food and Agriculture University, Konya, Türkiye
| |
Collapse
|
3
|
González SPR, Sandoval LMB, Giovanny TV. Effect of a probiotic mixture with lactic acid activity on productive and allometric indicators in broiler chickens. Vet World 2024; 17:1490-1496. [PMID: 39185053 PMCID: PMC11344101 DOI: 10.14202/vetworld.2024.1490-1496] [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: 03/09/2024] [Accepted: 06/07/2024] [Indexed: 08/27/2024] Open
Abstract
Background and Aim The feeding and sanitary conditions significantly influence the productivity of farm animals. This study aimed to assess the impact of a lactic acid-producing microbial additive on broiler chicken productivity. Materials and Methods A 42-day experimental period utilized 120 1-day-old Cobb 500 chicks with an average weight of 46 g. In groups of 30 each, the chicks were randomly assigned to four experimental designs. The following treatments were assessed: T1 without intervention (control), T2 with bacitracin at a concentration of 0.5 g/L, T3 with a 5% probiotic mixture (PM), and T4 with a 7.5% PM. The birds were fed the commercial balanced feed without anticoccidials daily, while vaccines were administered according to the recommended biosecurity plan by the commercial house. Drinking water was treated with PM containing lactobacilli, yeasts, and short-chain organic acids. Result In T4, a 7.5% PM resulted in a final weight of 2361.2 g (p < 0.05), a total weight gain of 1412.8 g (p < 0.05), and improved feed efficiency with a feed conversion of 2.00 (p < 0.05), during which feed intake was lower than in the other groups. Conclusion Microbial additives with lactic acid activity are a cost-effective and feasible solution for broiler chicken productivity.
Collapse
Affiliation(s)
- Sandra Paola Rodríguez González
- Veterinary Medicine and Zootechnics Program, Pedagogical and Technological University of Colombia, Tunja, Boyacá, Colombia
- GIBNA; Research Group in Biochemistry and Animal Nutrition, Tunja, Boyacá, Colombia
| | - Luis Miguel Borras Sandoval
- Veterinary Medicine and Zootechnics Program, Pedagogical and Technological University of Colombia, Tunja, Boyacá, Colombia
- GIDIMEVEZ Research Group in Veterinary Medicine and Zootechnics, Tunja, Boyacá, Colombia
| | - Torres Vidales Giovanny
- Veterinary Medicine and Zootechnics Program, Pedagogical and Technological University of Colombia, Tunja, Boyacá, Colombia
- GIBNA; Research Group in Biochemistry and Animal Nutrition, Tunja, Boyacá, Colombia
| |
Collapse
|
4
|
Zheng J, Ahmad AA, Yang C, Liang Z, Shen W, Liu J, Yan Z, Han J, Yang Y, Dong P, Lan X, Salekdeh GH, Ding X. Orally Administered Lactobacillus rhamnosus CY12 Alleviates DSS-Induced Colitis in Mice by Restoring the Intestinal Barrier and Inhibiting the TLR4-MyD88-NF-κB Pathway via Intestinal Microbiota Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38598717 DOI: 10.1021/acs.jafc.3c07279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Oral ingestion of probiotics is a promising approach to relieving inflammatory disease through regulating the gut microbiota. A newly discovered strain, Lactobacillus rhamnosus CY12 (LCY12), obtained from cattle-yak milk, displayed numerous probiotic properties. These included enhanced viability in low pH and bile environments, adhesion capabilities, and potent antimicrobial effects. The research aimed to explore the beneficial impacts of the novel LCY12 strain on colitis in mice induced by dextran sulfate sodium (DSS) and to elucidate the underlying molecular mechanisms. The results of the study showed that administration of LCY12 effectively helped to reduce the negative effects of DSS-induced body weight loss, disease activity index score, colon length shortening, loss of goblet cells, and overall histopathological scores in the intestines. Simultaneously, LCY12 administration significantly alleviated intestinal inflammation and safeguarded intestinal barrier integrity by enhancing IL-10 levels, while dampening IL-6, IL-1β, and TNF-α production. Additionally, LCY12 boosted the presence of tight junction proteins. Furthermore, LCY12 hindered the TLR4/MyD88/NF-κB signaling pathway by downregulating TLR4 and MyD88 expression, inactivating phosphorylated IκBα, and preventing translocation of NF-κB p65 from the cytoplasm to the nucleus. The LCY12 also increased specific intestinal microbial communities and short-chain fatty acid (SCFA) production. Altogether, LCY12 oral administration alleviated colitis induced with DSS in mice by improving intestinal barrier function and regulating inflammatory cytokines, SCFA production, and intestinal microbiota.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Anum Ali Ahmad
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH8 9YL, U.K
| | - Chen Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zeyi Liang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jing Liu
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zuoting Yan
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jianlin Han
- Livestock Genetics Program, International Livestock Research Institute (ILRI), 00100 Nairobi, Kenya
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Pengcheng Dong
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| |
Collapse
|
5
|
Akhgarjand C, Vahabi Z, Shab-Bidar S, Anoushirvani A, Djafarian K. The effects of probiotic supplements on oxidative stress and inflammation in subjects with mild and moderate Alzheimer's disease: a randomized, double-blind, placebo-controlled study. Inflammopharmacology 2024; 32:1413-1420. [PMID: 38319476 DOI: 10.1007/s10787-023-01427-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024]
Abstract
Through modulating effects on the gut-brain axis, probiotics are an effective adjuvant treatment for Alzheimer's disease (AD), one of our century's most important medical care challenges (Agahi et al. Front Neurol 9:662, 2018). This trial aimed to examine the effects of two different single-strain probiotics on oxidative stress and inflammation in patients with mild and moderate AD. This was a 12-week placebo-controlled, double-blind, randomized clinical trial performed on 90 patients with AD. Eligible patients were randomly assigned to two different interventions (Lacticaseibacillus rhamnosus HA-114 (7.5 × 109) or Bifidobacterium longum R0175 (7.5 × 109)) and a placebo group, supplemented twice daily. We used mixed-effect models to examine the probiotic's independent effects on clinical results. Significant improvements in serum inflammatory and oxidative stress markers were observed at the end of the trial (P < 0.05). Probiotic supplementation for 12 weeks had beneficial effects on oxidative stress, inflammation, quality of life, and physical activity in patients with mild and moderate AD.
Collapse
Affiliation(s)
- Camellia Akhgarjand
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Cognitive Neurology and Neuropsychiatry Division, Psychiatry Department, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran.
- Geriatric Department, Ziaeeian Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliarash Anoushirvani
- Department of Internal Medicine, School of Medicine, Firoozgar General Hospital, Iran University of Medical Science, Tehran, Iran
| | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Wang Z, Zhang Z, Shi Q, Liu S, Wu Q, Wang Z, Saiding E, Han J, Zhou J, Wang R, Su X. Whole genome sequencing analysis of Limosilactobacillus reuteri from the intestinal tract of mice recovering from ulcerative colitis and preliminary study on anti-inflammatory effects of its derived peptides. Arch Microbiol 2024; 206:140. [PMID: 38441642 DOI: 10.1007/s00203-024-03906-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/07/2024]
Abstract
Limosilactobacillus reuteri is an indigenous inhabitant of the animal gut known for its probiotic effects on the host. In our previous study, a large number of L. reuteri strains were isolated from the gastrointestinal tract of mice recovering from ulcerative colitis, from which we randomly selected L. reuteri RE225 for whole genome sequencing to explore its probiotic properties. The results of next-generation sequencing and third-generation single molecule sequencing showed that L. reuteri RE225 contained many genes encoding functional proteins associated with adhesion, anti-inflammatory and pathogen inhibition. And compared to other L. reuteri strains in NCBI, L. reuteri RE225 has unique gene families with probiotic functions. In order to further explore the probiotic effect of the L. reuteri RE225, the derived peptides were identified by LC-MS/MS, and the peptides with tumor necrosis factor-α binding ability were screened by reverse molecular docking and microscale thermophoresis. Finally, cell experiments demonstrated the anti-inflammatory ability of the peptides. Western blotting and qPCR analyses confirmed that the selected peptides might alleviate LPS-induced inflammation in NCM460 cells by inhibiting JAK2/STAT3 pathway activation.
Collapse
Affiliation(s)
- Ziyan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Zhixuan Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Qiuyue Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Songyi Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Qiaoli Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Ze Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Emilaguli Saiding
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Jiaojiao Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Jun Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China
| | - Rixin Wang
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China.
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China.
| | - Xiurong Su
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo, China.
- School of Marine Science, Ningbo University, 169 Qixing South Road, Ningbo, China.
| |
Collapse
|
7
|
Hu R, Yang T, Ai Q, Shi Y, Ji Y, Sun Q, Tong B, Chen J, Wang Z. Autoinducer-2 promotes the colonization of Lactobacillus rhamnosus GG to improve the intestinal barrier function in a neonatal mouse model of antibiotic-induced intestinal dysbiosis. J Transl Med 2024; 22:177. [PMID: 38369503 PMCID: PMC10874557 DOI: 10.1186/s12967-024-04991-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Human health is seriously threatened by antibiotic-induced intestinal disorders. Herein, we aimed to determine the effects of Autoinducer-2 (AI-2) combined with Lactobacillus rhamnosus GG (LGG) on the intestinal barrier function of antibiotic-induced intestinal dysbiosis neonatal mice. METHODS An antibiotic-induced intestinal dysbiosis neonatal mouse model was created using antibiotic cocktails, and the model mice were randomized into the control, AI-2, LGG, and LGG + AI-2 groups. Intestinal short-chain fatty acids and AI-2 concentrations were detected by mass spectrometry and chemiluminescence, respectively. The community composition of the gut microbiota was analyzed using 16S rDNA sequencing, and biofilm thickness and bacterial adhesion in the colon were assessed using scanning electron microscopy. Transcriptome RNA sequencing of intestinal tissues was performed, and the mRNA and protein levels of HCAR2 (hydroxycarboxylic acid receptor 2), claudin3, and claudin4 in intestinal tissues were determined using quantitative real-time reverse transcription PCR and western blotting. The levels of inflammatory factors in intestinal tissues were evaluated using enzyme-linked immunosorbent assays (ELISAs). D-ribose, an inhibitor of AI-2, was used to treat Caco-2 cells in vitro. RESULTS Compared with the control, AI-2, and LGG groups, the LGG + AI-2 group showed increased levels of intestinal AI-2 and proportions of Firmicutes and Lacticaseibacillus, but a reduced fraction of Proteobacteria. Specifically, the LGG + AI-2 group had considerably more biofilms and LGG on the colon surface than those of other three groups. Meanwhile, the combination of AI-2 and LGG markedly increased the concentration of butyric acid and promoted Hcar2, claudin3 and claudin4 expression levels compared with supplementation with LGG or AI-2 alone. The ELISAs revealed a significantly higher tumor necrosis factor alpha (TNF-α) level in the control group than in the LGG and LGG + AI-2 groups, whereas the interleukin 10 (IL-10) level was significantly higher in the LGG + AI-2 group than in the other three groups. In vitro, D-ribose treatment dramatically suppressed the increased levels of Hcar2, claudin3, and claudin4 in Caco-2 cells induced by AI-2 + LGG. CONCLUSIONS AI-2 promotes the colonization of LGG and biofilm formation to improve intestinal barrier function in an antibiotic-induced intestinal dysbiosis neonatal mouse model.
Collapse
Affiliation(s)
- Riqiang Hu
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ting Yang
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Qing Ai
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanchun Ji
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Sun
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Tong
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Jie Chen
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| | - Zhengli Wang
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Jiangxi Hospital Affiliated Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Su X, Menghe B, Zhang H, Liu W. In Vitro Evaluation of Intestinal Transport and High-Density Fermentation of Lactobacillus acidophilus. Metabolites 2023; 13:1077. [PMID: 37887401 PMCID: PMC10609339 DOI: 10.3390/metabo13101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023] Open
Abstract
Lactobacillus acidophilus strains have limiting factors such as low cell density and complex nutritional requirements in industrial production, which greatly restricts their industrial application. In this study, fermentation conditions for L. acidophilus were optimized and transcriptomic analysis used to understand growth mechanisms under high-density fermentation conditions. We found that L. acidophilus IMAU81186 has strong tolerance to gastrointestinal juice. In addition, its optimal culture conditions were 3% inoculum (v/v); culture temperature 37 °C; initial pH 6.5; and medium composition of 30.18 g/L glucose, 37.35 g/L soybean peptone, 18.68 g/L fish peptone, 2.46 g/L sodium citrate, 6.125 g/L sodium acetate, 2.46 g/L K2HPO4, 0.4 g/L MgSO4·7H2O,0.04 g/L MnSO4·5H2O, 0.01 g/L serine, and 0.3 g/L uracil. After optimization, viable counts of IMAU81186 increased by 7.03 times. Differentially expressed genes in IMAU81186 were analyzed at different growth stages using transcriptomics. We found that a single carbon source had limitations in improving the biomass of the strain, and terP and bfrA were significantly down-regulated in the logarithmic growth period, which may be due to the lack of extracellular sucrose. After optimizing the carbon source, we found that adding 12 g/L sucrose to the culture medium significantly increased cell density.
Collapse
Affiliation(s)
- Xin Su
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.S.); (B.M.); (H.Z.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
- Collaborative Innovative Center of Ministry of Education for Lactic Acid Bacteria and Fermented Dairy Products, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Bilige Menghe
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.S.); (B.M.); (H.Z.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
- Collaborative Innovative Center of Ministry of Education for Lactic Acid Bacteria and Fermented Dairy Products, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.S.); (B.M.); (H.Z.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
- Collaborative Innovative Center of Ministry of Education for Lactic Acid Bacteria and Fermented Dairy Products, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenjun Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.S.); (B.M.); (H.Z.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
- Collaborative Innovative Center of Ministry of Education for Lactic Acid Bacteria and Fermented Dairy Products, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
9
|
Ismael M, Qayyum N, Gu Y, Zhezhe Y, Cui Y, Zhang Y, Lü X. Protective effect of plantaricin bio-LP1 bacteriocin on multidrug-resistance Escherichia Coli infection by alleviate the inflammation and modulate of gut-microbiota in BALB/c mice model. Int J Biol Macromol 2023; 246:125700. [PMID: 37414312 DOI: 10.1016/j.ijbiomac.2023.125700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
The rapid spread of multidrug-resistant pathogens with the low efficacy of common antibiotics for humans and animals in its clinical therapeutics are a global health concern. Therefore, there is a need to develop new treatment strategies to control them clinically. The study aimed to evaluate the effects of Plantaricin Bio-LP1 bacteriocin produced from Lactiplantibacillus plantarum NWAFU-BIO-BS29 to alleviate the inflammation caused by multidrug-resistance Escherichia Coli (MDR-E. coli) infection in BALB/c mice-model. The focus was given on aspects linked to the mechanism of the immune response. Results indicated that Bio-LP1 had highly promising effects on partially ameliorating MDR-E. coli infection by reducing the inflammatory response through inhibiting the overexpression of proinflammatory-cytokines such as secretion of tumor necrosis factor (TNF-α) and interleukin (IL-6 and IL-β) and strongly regulated theTLR4 signaling-pathway. Additionally, avoided the villous destruct, colon length shortening, loss of intestinal barrier integrity, and increased disease activity index. Furthermore, significantly increased the relative abundance of beneficial-intestinal-bacteria including Ligilactobacillus, Enterorhabdus, Pervotellaceae, etc. Finally, improved the intestinal mucosal barrier to alleviate the pathological damages and promote the production of short-chain fatty acids (SCFAs) a source of energy for the proliferation. In conclusion, plantaricin Bio-LP1 bacteriocin can be considered a safe alternative to antibiotics against MDR-E. coli-induced intestinal inflammation.
Collapse
Affiliation(s)
- Mohamedelfatieh Ismael
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; Sudanese Standard and Metrology Organization, Khartoum, 13573, Sudan
| | - Nageena Qayyum
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yaxin Gu
- College of Food Science, China Agricultural University, Beijing, China
| | - Yu Zhezhe
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yanlong Cui
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yu Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
10
|
Suriano ES, Souza MDM, Kobata CM, Santos FHY, Mimica MJ. Efficacy of an adjuvant Lactobacillus rhamnosus formula in improving skin lesions as assessed by PASI in patients with plaque psoriasis from a university-affiliated, tertiary-referral hospital in São Paulo (Brazil): a parallel, double-blind, randomized clinical trial. Arch Dermatol Res 2023; 315:1621-1629. [PMID: 36757438 DOI: 10.1007/s00403-023-02553-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/03/2022] [Accepted: 01/22/2023] [Indexed: 02/10/2023]
Abstract
Psoriasis is an inflammatory disease of the skin, characterized by erythematous plaques. It is rather common, affecting 2-4% of the population in western countries. Psoriasis' etiology encompasses both genetic and environmental factors. Evidence suggests that the latter reflect the importance of changes in the microbiome for developing the disease. Thus, it is hypothesized that gut microbiome manipulation may arise as a way of treating psoriasis. However, few trials assessed the use of probiotics in psoriasis, although promising results were detected in small studies. Our objective was to assess the efficacy of adjuvant probiotics (Lactobacillus rhamnosus) in treating plaque psoriasis patients. This was a randomized, parallel, placebo-controlled, double-blind trial with two arms: experimental (n = 50) and control (n = 53). Inclusion of subjects and data gathering lasted from November 2020 to August 2021. Subjects were consecutive plaque psoriasis patients under regular follow-up in the Dermatology unit of a university-affiliated, tertiary-referral hospital in São Paulo (Brazil). Eligibility criteria included being over 18 years old, having plaque psoriasis and not having other skin diseases, neoplasms nor systemic inflammatory diseases. Subjects received standard-of-care plus probiotics (Lactobacillus rhamnosus formula). Controls received standard-of-care plus placebo. Primary outcome was skin lesion improvement as assessed by psoriasis area of severity index (PASI) at six months. Secondary outcome was quality-of-life as assessed by dermatology life quality index (DLQI) at six months. Regarding within-group analyses, changes in both PASI and DLQI were non-significant for the experimental group (mean PASI decreased by 1.58, p = 0.105, and mean DLQI increased by 0.05, p = 0.873) and significant for controls (mean PASI decreased by 1.90, p = 0.019, and mean DLQI decreased by 3.33, p = 0.031). Between-group analyses returned non-significant results (p = 0.620). Our findings do not support the hypothesis that gut microbiome modulation via ingestion of Lactobacillus rhamnosus produces clinical improvement in psoriasis patients. Further research is encouraged.Trial registration: Retrospectively registered at the Brazilian Clinical Trials Registry (RBR-8js7t83) on 08/02/2022.
Collapse
Affiliation(s)
- Enrico Stefano Suriano
- Santa Casa de São Paulo Medical School, R. Dr. Cesário Mota Júnior, 61 Vila Buarque, São Paulo, SP, 01221020, Brazil.
- Rua Corrientes, 135, Apartment 42W, Lapa, São Paulo, SP, 05076010, Brazil.
| | - Marília Diogo Moço Souza
- Santa Casa de São Paulo Medical School, R. Dr. Cesário Mota Júnior, 61 Vila Buarque, São Paulo, SP, 01221020, Brazil
| | - Clarice Marie Kobata
- Santa Casa de São Paulo Medical School, R. Dr. Cesário Mota Júnior, 61 Vila Buarque, São Paulo, SP, 01221020, Brazil
| | - Felipe Henrique Yazawa Santos
- Santa Casa de São Paulo Medical School, R. Dr. Cesário Mota Júnior, 61 Vila Buarque, São Paulo, SP, 01221020, Brazil
| | - Marcelo Jenné Mimica
- Santa Casa de São Paulo Medical School, R. Dr. Cesário Mota Júnior, 61 Vila Buarque, São Paulo, SP, 01221020, Brazil
| |
Collapse
|
11
|
Trindade LM, Torres L, Matos ID, Miranda VC, de Jesus LCL, Cavalcante G, de Souza Oliveira JJ, Cassali GD, Mancha-Agresti P, de Carvalho Azevedo VA, Maioli TU, Cardoso VN, Martins FDS, de Vasconcelos Generoso S. Paraprobiotic Lacticaseibacillus rhamnosus Protects Intestinal Damage in an Experimental Murine Model of Mucositis. Probiotics Antimicrob Proteins 2023; 15:338-350. [PMID: 34524605 DOI: 10.1007/s12602-021-09842-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/17/2022]
Abstract
Intestinal mucositis (IM) is a common side effect resulting from cancer treatment. However, the management so far has not been very effective. In the last years, the role of the gut microbiota in the development and severity of mucositis has been studied. Therefore, the use of probiotics and paraprobiotics could have a potential therapeutic effect on IM. The aim of our study was to investigate the impact of the administration of Lacticaseibacillus rhamnosus (L. rhamnosus) CGMCC1.3724 and the paraprobiotic on IM in mice. For 13 days, male Balb/c mice were divided into six groups: control (CTL) and mucositis (MUC)/0.1 mL of saline; CTL LrV and MUC LrV/0.1 mL of 108 CFU of viable Lr; CTL LrI and MUC LrI/0.1 mL of 108 CFU of inactivated Lr. On the 10th day, mice from the MUC, MUC LrV, and MUC LrI groups received an intraperitoneal injection (300 mg/kg) of 5-fluorouracil to induce mucositis. The results showed that the administration of the chemotherapeutic agent increased the weight loss and intestinal permeability of the animals in the MUC and MUC LrV groups. However, administration of paraprobiotic reduced weight loss and maintained PI at physiological levels. The paraprobiotic also preserved the villi and intestinal crypts, reduced the inflammatory infiltrate, and increased the mucus secretion, Muc2 gene expression, and Treg cells frequency.
Collapse
Affiliation(s)
- Luísa Martins Trindade
- Programa de Pós-Graduação Em Ciência de Alimentos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lícia Torres
- Programa de Pós-Graduação Em Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabel David Matos
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian Correia Miranda
- Programa de Pós-Graduação Em Microbiologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gregório Cavalcante
- Programa de Pós-Graduação Em Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Geovanni Dantas Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flaviano Dos Santos Martins
- Programa de Pós-Graduação Em Microbiologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone de Vasconcelos Generoso
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
12
|
Two doses of Lactobacillus induced different microbiota profiles and serum immune indices in pigs. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023] Open
|
13
|
Chen L, Dai M, Zuo W, Dai Y, Yang Q, Yu S, Huang M, Liu H. NF-κB p65 and SETDB1 expedite lipopolysaccharide-induced intestinal inflammation in mice by inducing IRF7/NLR-dependent macrophage M1 polarization. Int Immunopharmacol 2023; 115:109554. [PMID: 36580757 DOI: 10.1016/j.intimp.2022.109554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/04/2022] [Accepted: 12/04/2022] [Indexed: 12/28/2022]
Abstract
Macrophages exhibit distinct phenotypes that are pro-inflammatory (M1) or anti-inflammatory (M2) in response to inflammation. In this study, we tried to identify the roles and mechanisms of interferon regulatory factor 7 (IRF7) in modulating the phenotypes of macrophages in lipopolysaccharide (LPS)-induced intestinal inflammation. The mouse model of intestinal inflammation was induced by lipopolysaccharide (LPS), and mouse bone marrow-derived macrophages (BMDMs) and mouse intestinal epithelial cells were selected for experimental verification in vitro. Results demonstrated that IRF7 was highly expressed in the mouse model of intestinal inflammation, while IRF7 deficiency repressed macrophage M1 polarization and attenuated intestinal inflammation in mice. p65 and SET domain bifurcated 1 (SETDB1) synergistically promoted histone 3 lysine 4 trimethylation (H3K4me3) methylation to elevate IRF7 expression, which activated the Nod-like receptor (NLR) pathway to induce macrophage M1 polarization. Through this mechanism, IRF7 in BMDMs functioned to accelerate intestinal epithelial cell apoptosis and their release of pro-inflammatory proteins. Furthermore, the promoting effect of p65 and SETDB1 on LPS-induced intestinal inflammation was validated in vivo. To sum up, NF-κB p65 and SETDB1 facilitated IRF7-mediated macrophage M1 polarization, thereby aggravating the LPS-induced intestinal inflammation. Hence, this study highlights the appealing value of these factors as anti-inflammatory targets.
Collapse
Affiliation(s)
- Li Chen
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Maolin Dai
- Department of Anesthesia, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Wei Zuo
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Yongyu Dai
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Qiqi Yang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Shuangjiang Yu
- Department of Neurosurgery, The First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing 400038, PR China
| | - Min Huang
- Department of Digestion, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, PR China
| | - Hao Liu
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China.
| |
Collapse
|
14
|
Li Z, Zhang S, Xu L, Fang X, Wan Y, Yu D, Guo Y. A tetrapeptide from maize combined with probiotics exerted strong anti-inflammatory effects and modulated gut microbiota in DSS-induced colitis mice. Food Funct 2022; 13:12602-12618. [PMID: 36373867 DOI: 10.1039/d2fo02678c] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by recurrent gastrointestinal inflammation caused by abnormal immune response, and patients usually have intestinal flora imbalance. At present, the pathogenesis of UC is not well understood, and it appears that there is chronic activation of the immune and inflammatory cascade in genetically susceptible individuals. Some food supplements such as specific peptides and probiotics have been investigated and shown the potential for the treatment of UC. The purpose of this study is to investigate the therapeutic effect and potential mechanism of tetrapeptide from maize (TPM) and probiotic treatment on dextran sulfate sodium (DSS)-induced UC in C57BL/6J mice. Our results indicated that the therapeutic effects of TPM and probiotics are positively associated with a reduction in pro-inflammatory cytokine levels and restoration of the gut microbiota. Treatment with TPM or probiotics effectively alleviated the adverse effects of UC, including weight loss, shortened colon length, and colon and kidney tissue damage in mice. Additionally, both TPM and probiotics significantly reduced pro-inflammatory cytokine levels and oxidative stress in UC mice, and the effect was more pronounced when both were used together. Moreover, co-treatment with TPM and probiotics increased the diversity of gut microbes in UC mice, reduced the ratio of Firmicutes to Bacteroidetes (F/B) and increased the abundance of bacterial species, including Muribaculaceae, Alistipes, Ligilactobacillus and Lactobacillus, and has been shown to be beneficial for a variety of pathological conditions.
Collapse
Affiliation(s)
- Zhiguo Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| | - Shan Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| | - Li Xu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| | - Youzhong Wan
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China 130033, P. R. China
| | - Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| | - Yi Guo
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China.
| |
Collapse
|
15
|
Xin WG, Li XD, Lin YC, Jiang YH, Xu MY, Zhang QL, Wang F, Lin LB. Whole genome analysis of host-associated lactobacillus salivarius and the effects on hepatic antioxidant enzymes and gut microorganisms of Sinocyclocheilus grahami. Front Microbiol 2022; 13:1014970. [PMID: 36386721 PMCID: PMC9648147 DOI: 10.3389/fmicb.2022.1014970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 09/29/2023] Open
Abstract
As a fish unique to Yunnan Province in China, Sinocyclocheilus grahami hosts abundant potential probiotic resources in its intestinal tract. However, the genomic characteristics of the probiotic potential bacteria in its intestine and their effects on S. grahami have not yet been established. In this study, we investigated the functional genomics and host response of a strain, Lactobacillus salivarius S01, isolated from the intestine of S. grahami (bred in captivity). The results revealed that the total length of the genome was 1,737,623 bp (GC content, 33.09%), comprised of 1895 genes, including 22 rRNA operons and 78 transfer RNA genes. Three clusters of antibacterial substances related genes were identified using antiSMASH and BAGEL4 database predictions. In addition, manual examination confirmed the presence of functional genes related to stress resistance, adhesion, immunity, and other genes responsible for probiotic potential in the genome of L. salivarius S01. Subsequently, the probiotic effect of L. salivarius S01 was investigated in vivo by feeding S. grahami a diet with bacterial supplementation. The results showed that potential probiotic supplementation increased the activity of antioxidant enzymes (SOD, CAT, and POD) in the hepar and reduced oxidative damage (MDA). Furthermore, the gut microbial community and diversity of S. grahami from different treatment groups were compared using high-throughput sequencing. The diversity index of the gut microbial community in the group supplemented with potential probiotics was higher than that in the control group, indicating that supplementation with potential probiotics increased gut microbial diversity. At the phylum level, the abundance of Proteobacteria decreased with potential probiotic supplementation, while the abundance of Firmicutes, Actinobacteriota, and Bacteroidota increased. At the genus level, there was a decrease in the abundance of the pathogenic bacterium Aeromonas and an increase in the abundance of the potential probiotic bacterium Bifidobacterium. The results of this study suggest that L. salivarius S01 is a promising potential probiotic candidate that provides multiple benefits for the microbiome of S. grahami.
Collapse
Affiliation(s)
- Wei-Gang Xin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Xin-Dong Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Yi-Cen Lin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Yu-Hang Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Mei-Yu Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Qi-Lin Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Feng Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| | - Lian-Bing Lin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan Kunming, China
- Engineering Research Center for Replacement Technology of Feed Antibiotics of Yunnan College, Yunnan, Kunming, China
| |
Collapse
|
16
|
Chen X, Gao F, Zhang J. Lactobacillus complex capsules ameliorate aspirin-related small intestinal mucosal injury: a prospective, randomized, controlled clinical trial. Scand J Gastroenterol 2022; 57:1195-1201. [PMID: 35534443 DOI: 10.1080/00365521.2022.2073184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Aspirin can reduce cardiovascular disease risk; however, it can increase the risk of gastrointestinal injury. Lactobacilli have some protective effects; however, there are few studies on their effects on humans. This study investigates the effects of Lactobacillus complex capsule treatment on the aspirin-related small intestinal mucosal injury. METHODS This single-center, prospective, randomized controlled clinical trial included 69 patients using enteric-coated aspirin for >1 month between May and December 2019. After baseline magnetically controlled capsule endoscopy (MCCE), patients with aspirin-related small intestinal mucosal injury were randomly assigned (1:1) to receive enteric-coated aspirin and Lactobacillus complex capsules containing a combination of Lactobacillus rhamnosus I, Lactobacillus rhamnosus II, and Enterococcus faecium (probiotics group) or enteric-coated aspirin only (control group) for 2 months. After treatment, the patient underwent MCCE again. The primary outcome was the change in small intestinal mucosal injury scores from baseline to post-intervention. RESULTS Twenty-five patients in the probiotics group and 28 in the control group completed the trial. The decrease in small intestinal mucosal injury scores from baseline to post-intervention was significantly greater in the probiotics group than that in the control group (p < .001). The improvement rates of red spots and erosions in the probiotics group were higher compared with the control group (p = .027 and .022, respectively), and the improvement rate of small intestinal ulcers in the probiotics group was 75.0%; however, there was no improvement in the control group. CONCLUSION Lactobacillus complex capsules can ameliorate aspirin-related small intestinal mucosal injury.
Collapse
Affiliation(s)
- Xue Chen
- Department of Gastroenterology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Feng Gao
- Department of Gastroenterology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jie Zhang
- Department of Gastroenterology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Zheng J, Ahmad AA, Yang Y, Liang Z, Shen W, Feng M, Shen J, Lan X, Ding X. Lactobacillus rhamnosus CY12 Enhances Intestinal Barrier Function by Regulating Tight Junction Protein Expression, Oxidative Stress, and Inflammation Response in Lipopolysaccharide-Induced Caco-2 Cells. Int J Mol Sci 2022; 23:ijms231911162. [PMID: 36232464 PMCID: PMC9569798 DOI: 10.3390/ijms231911162] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). The objectives of this study were to assess whether the Lactobacillus rhamnosus CY12 could alleviate oxidative stress, inflammation, and the disruption of tight junction (TJ) barrier functions induced by lipopolysaccharide (LPS), and therefore to explore the potential underlying molecular mechanisms. Our results showed that LPS-induced Cancer coli-2 (Caco-2) cells significantly increased the levels of reactive oxygen species (ROS), lactate dehydrogenase, inflammatory cytokines interleukin-1β, interleukin-6, interleukin-8, and tumor necrosis factor-α (IL-1β, IL-6, IL-8, and TNF-α), and the cell apoptosis rate while decreasing the levels of TJ proteins occludin, zonula occludens-1 (ZO-1), and claudin and antioxidant enzymes, such as catalase, superoxide dismutase, and glutathione peroxidase(CAT, SOD, and GSH-Px) (p < 0.05). However, Lactobacillus rhamnosus CY12 could relieve cytotoxicity, apoptosis, oxidative stress, and pro-inflammatory cytokine expressions, and also inhibit the Toll-like receptor 4/nuclear factor kappa-B(TLR4/NF-κB) signaling pathway. Furthermore, the gene expression of antioxidant enzymes, as well as the mRNA and protein expressions of TJ proteins, was improved. Particularly, the concentration of 108 cfu/mL significantly prevented the inflammatory injury induced by LPS in Caco-2 cells (p < 0.05). These findings support a potential application of Lactobacillus rhamnosus CY12 as a probiotic to prevent LPS-induced intestinal injury and treat intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Anum Ali Ahmad
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- State Key Laboratory of Grassland Agro-Ecosystems, School of Life Sciences, Lanzhou University, Lanzhou 730050, China
| | - Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zeyi Liang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Min Feng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jiahao Shen
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| | - Xuezhi Ding
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| |
Collapse
|
18
|
Limnospira indica PCC 8005 or Lacticaseibacillus rhamnosus GG Dietary Supplementation Modulate the Gut Microbiome in Mice. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2030049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
While dietary supplements can have beneficial effects on the health of the intestine, these effects can come with unresolved issues in terms of therapeutic efficacy and mechanisms of action. In this study, the model probiotic Lacticaseibacillus rhamnosus GG ATCC 53103 and the anciently used dietary supplement Limnospira indica strain PCC 8005 were compared for their effects on murine intestinal ecology. Healthy male mice received either saline or suspensions of living cells of L. indica PCC 8005 or L. rhamnosus GG daily along a two-week intervention period, followed by a two-week washout period. Both bacteria-based solutions appeared able to transiently shift the microbial community, which were characterized by a higher relative abundance of members of the butyrate producing Lachnospiraceae and Porphyromonadaceae families.
Collapse
|
19
|
Rahim MA, Seo H, Kim S, Tajdozian H, Barman I, Lee Y, Lee S, Song HY. In vitro anti-tuberculosis effect of probiotic Lacticaseibacillus rhamnosus PMC203 isolated from vaginal microbiota. Sci Rep 2022; 12:8290. [PMID: 35585245 PMCID: PMC9116076 DOI: 10.1038/s41598-022-12413-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/05/2022] [Indexed: 11/09/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb), the etiological agent of tuberculosis (TB), poses a severe challenge for public health and remains the number one cause of death as a single infectious agent. There are 10 million active cases of TB per year with 1.5 million deaths, and 2-3 billion people are estimated to harbor latent M. tb infection. Moreover, the emergence of multi-drug-resistant (MDR), extremely-drug-resistant (XDR), and the recent totally drug-resistant (TDR) M. tb is becoming a global issue that has fueled the need to find new drugs different from existing regimens. In these circumstances, probiotics can be a potential choice, so we focused on developing them as an anti-tuberculosis drug candidate. Here, we report the anti-tubercular activities of Lacticaseibacillus rhamnosus PMC203 isolated from the vaginal microbiota of healthy women. PMC203 exhibited a promising intracellular killing effect against both drug-sensitive and resistant M. tb infected murine macrophage cell line RAW 264.7 without showing any cytotoxicity. Additionally, it also inhibited the growth of M. tb under broth culture medium. PMC203 did not cause weight change or specific clinical symptoms in a 2-week repeated oral administration toxicity test in a guinea pig model. Here, we also found that PMC203 induces autophagy in a dose dependent manner by increasing the signal of well-known autophagy gene markers, suggesting a possible intracellular killing mechanism.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea.,Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Chungnam, Korea
| | - Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea
| | - Hanieh Tajdozian
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea.,Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Chungnam, Korea
| | - Indrajeet Barman
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea.,Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Chungnam, Korea
| | - Youngkyoung Lee
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea.,Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Chungnam, Korea
| | - Saebim Lee
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea
| | - Ho-Yeon Song
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Chungnam, Korea. .,Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Chungnam, Korea.
| |
Collapse
|
20
|
Ge L, Liu D, Mao X, Liu S, Guo J, Hou L, Chen X, Huang K. Low Dose of Deoxynivalenol Aggravates Intestinal Inflammation and Barrier Dysfunction Induced by Enterotoxigenic Escherichia coli Infection through Activating Macroautophagy/NLRP3 Inflammasomes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3009-3022. [PMID: 35201764 DOI: 10.1021/acs.jafc.1c07834] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The toxicity of deoxynivalenol (DON) in healthy humans and animals has been extensively studied. However, whether the natural-low-dose DON is scatheless under unhealthy conditions, especially intestinal injury, is unknown. Infection of enterotoxigenic Escherichia coli (ETEC) is a classical intestinal injury model. In this study, we explored the effects of low-dose DON on intestinal injury induced by the ETEC infection and the underlying mechanism in piglets, mice, and IPEC-J2 monolayer cells. Results showed that significant growth slowdown, severe diarrhea, and intestinal damage, bacterial multiplication, and translocation were observed in the experimental group (low-dose DON, 0.75 mg/kg in feed for piglets, and 1 mg/kg body weight for mice, combined with the ETEC infection). Meanwhile, more aggressive intestinal inflammation and barrier dysfunction were observed in animals and IPEC-J2 monolayer cells. Higher expression levels of NLRP3 inflammasome and LC3B were observed in jejunum and IPEC-J2 in the experimental group. After treatment with NLRP3 or caspase1 inhibitors, excessive intestinal inflammation rather than barrier dysfunction in the experimental group was limited. CRISPR-Cas9-mediated knockout of LC3B alleviated intestinal inflammation and barrier dysfunction and also inhibited NLRP3 inflammasome. In conclusion, a low dose of DON aggravates intestinal inflammation and barrier dysfunction induced by the ETEC infection by activating macroautophagy and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xinru Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Junyan Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Lili Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| |
Collapse
|
21
|
Kim WJ, Hyun JH, Lee NK, Paik HD. Protective Effects of a Novel Lactobacillus brevis Strain with Probiotic Characteristics against Staphylococcus aureus Lipoteichoic Acid-Induced Intestinal Inflammatory Response. J Microbiol Biotechnol 2022; 32:205-211. [PMID: 34750285 PMCID: PMC9628842 DOI: 10.4014/jmb.2110.10034] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/15/2022]
Abstract
Probiotics can effectively modulate host immune responses and prevent gastrointestinal diseases. The objective of this study was to investigate the probiotic characteristics of Lactobacillus brevis KU15152 isolated from kimchi and its protective potential against intestinal inflammation induced by Staphylococcus aureus lipoteichoic acid (aLTA). L. brevis KU15152 exhibited a high survival rate in artificial gastric and bile environments. Additionally, the adhesion capability of the strain to HT-29 cells was higher than that of L. rhamnosus GG. L. brevis KU15152 did not produce harmful enzymes, such as β-glucuronidase, indicating that it could be used as a potential probiotic. The anti-inflammatory potential of L. brevis KU15152 was determined in HT-29 cells. Treatment with L. brevis KU15152 suppressed the production of interleukin-8 without inducing significant cytotoxicity. The downregulatory effects of L. brevis KU15152 were involved in the suppression of nuclear factor-kappa B activation mediated by the extracellular signal-regulated kinase and Akt signaling pathways. Collectively, these data suggest that L. brevis KU15152 can be used in developing therapeutic and prophylactic products to manage and treat aLTA-induced intestinal damage.
Collapse
Affiliation(s)
- Won-Ju Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Jun-Hyun Hyun
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea,Corresponding author Phone: +82-2-2049-6011 E-mail:
| |
Collapse
|
22
|
Zheng J, Du M, Jiang W, Zhang J, Shen W, Ma X, Liang Z, Shen J, Wu X, Ding X. In Vitro Probiotic Characteristics and Whole Genome Sequence Analysis of Lactobacillus Strains Isolated from Cattle-Yak Milk. BIOLOGY 2021; 11:44. [PMID: 35053042 PMCID: PMC8772927 DOI: 10.3390/biology11010044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/14/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022]
Abstract
Cattle-yak milk is an important raw material and an indispensable source of high-quality food for local farmers and herdsmen to produce ghee, milk residue, yogurt, and other dairy products. In this study, Lactobacillus strains were isolated from cattle-yak milk for potential probiotic candidates using a series of in vitro tests, including probiotic characterization and safety evaluation (antibiotic susceptibility and hemolytic ability). The results found that the Lactobacillus rhamnosus CY12 strain showed a high survival rate in bile salts, under acid conditions, and in the gastrointestinal juice environment, as well as showing high antimicrobial activity and adhesive potential. The safety evaluation showed that all strains were considered non-hemolytic. In addition, the whole-genome sequencing indicated that the strain CY12 spanned 2,506,167 bp, with an average length of 881 bp; the GC content in the gene region (%) was 47.35, contained 1347 protein-coding sequences, and accounted for 85.72% of the genome. The genome annotation showed that genes mainly focused on the immune system process, metabolic process, carbohydrate utilization, carbon metabolism, galactose metabolism, and biological adhesion, etc. This study revealed that the Lactobacillus rhamnosus CY12 strain might be an excellent potential probiotic in the development of feed additives for animals and has the ability to promote health.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
| | - Mei Du
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
| | - Wei Jiang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (W.J.); (W.S.); (X.M.)
| | - Jianbo Zhang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (W.J.); (W.S.); (X.M.)
| | - Xiaoyu Ma
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (W.J.); (W.S.); (X.M.)
| | - Zeyi Liang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
| | - Jiahao Shen
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
| | - Xiaohu Wu
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (W.J.); (W.S.); (X.M.)
| | - Xuezhi Ding
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.Z.); (M.D.); (J.Z.); (Z.L.); (J.S.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (W.J.); (W.S.); (X.M.)
| |
Collapse
|
23
|
Yang M, Zheng J, Zong X, Yang X, Zhang Y, Man C, Jiang Y. Preventive Effect and Molecular Mechanism of Lactobacillus rhamnosus JL1 on Food-Borne Obesity in Mice. Nutrients 2021; 13:3989. [PMID: 34836242 PMCID: PMC8621931 DOI: 10.3390/nu13113989] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/22/2023] Open
Abstract
Probiotics can prevent obesity and related metabolic complications. In our study, the protective effect and molecular mechanism of Lactobacillus rhamnosus JL1 (separated from the feces of healthy infants) on high-fat diet mice were investigated. After 10 weeks of dietary intervention with L. rhamnosus JL1 intervention, the body weight of the JL1 group (23.78 g) was significantly lower than that of the HFD group (26.59 g, p < 0.05) and the liver index was reduced. Serum biochemical analysis showed that the TC, TG and LDL-C contents of JL1 group mice were significantly decreased (p < 0.05). Histological images of the mice livers showed that the degree of lipid action and damage of hepatic cells were improved. L. rhamnosus JL1 activated the AMPK pathway, and reduced the gene expression of PPAR-γ, LXR-α and SREBP-1C. In addition, the protein expression of PPAR-γ and LXR-α were reduced. After dietary intervention with L. rhamnosus JL1, the concentration of acetic acid, propionic acid, and butyric acid were increased significantly, especially the concentration of butyric acid, which was 63.16% higher than that of the HFD group (p < 0.05). In conclusion, this study provided a theoretical reference for the development and application of probiotics derived from healthy infant feces in health products and functional foods.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China; (M.Y.); (J.Z.); (X.Z.); (X.Y.); (Y.Z.); (C.M.)
| |
Collapse
|
24
|
Zhang L, Ma H, Kulyar MFEA, Pan H, Li K, Li A, Mo Q, Wang Y, Dong H, Bao Y, Li J. Complete genome analysis of Lactobacillus fermentum YLF016 and its probiotic characteristics. Microb Pathog 2021; 162:105212. [PMID: 34597776 DOI: 10.1016/j.micpath.2021.105212] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
Lactobacillus fermentum (L. fermentum) YLF016 is a well-characterized probiotic with several favorable characteristics. This study aimed to analyze the probiotic characteristics of L. fermentum and uncover the genes implicated in its potential probiotic ability on the base of its genomics features. The complete genome of L. fermentum YLF016 was found to have a circular chromosome of 2,094,354 bp, and 51.46% G + C content without any plasmid. Its chromosome contained 2,130 predicted protein-encoding genes, 58 tRNA, and 15 rRNA-encoding genes. Also, it was found to have many other probiotic properties, such as a high survival rate in the gastrointestinal tract with strong adherence to intestinal cells, antibacterial activity against pathogens, and antioxidant activity. Moreover, the genome sequence analysis demonstrated specific genes coding for carbon metabolism pathway, genetic adaption, stress resistance, and adhesive ability. Further analysis revealed its non-hemolytic activity and its non-functional ability of virulence factors. In conclusion, L. fermentum YLF016 possesses many valuable probiotic properties that refer to its potential probiotic ability.
Collapse
Affiliation(s)
- Lihong Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Hongcai Ma
- Tibet Livestock Research Institute, Tibet Academy of Agriculture And Animal Science, Lhasa 850009, Tibet, People's Republic of China
| | | | - Huachun Pan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kewei Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaping Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Hailong Dong
- Laboratory of Detection and Monitoring of Highland Animal Disease, Tibet Agricultural and Animal Husbandry University, Linzhi, 860000, People's Republic of China
| | - Yuhua Bao
- Tibet Biological Pharmaceutical Factory, Lhasa 850009, People's Republic of China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China; Laboratory of Detection and Monitoring of Highland Animal Disease, Tibet Agricultural and Animal Husbandry University, Linzhi, 860000, People's Republic of China.
| |
Collapse
|
25
|
Zhao L, Xie Q, Etareri Evivie S, Liu D, Dong J, Ping L, Liu F, Li B, Huo G. Bifidobacterium dentium N8 with potential probiotic characteristics prevents LPS-induced intestinal barrier injury by alleviating the inflammatory response and regulating the tight junction in Caco-2 cell monolayers. Food Funct 2021; 12:7171-7184. [PMID: 34269367 DOI: 10.1039/d1fo01164b] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). This study aimed to investigate the potential mechanism behind the protective effects of B. dentium N8 on the intestinal barrier using the lipopolysaccharide (LPS)-induced Caco-2 cells model. Our probiotic validation results showed that B. dentium N8 had a higher adhesion ability and a more substantial inhibition effect on Escherichia coli ATCC 25922 adhesion to HT-29 cells. Regarding the epithelial integrity, B. dentium N8 significantly increased the trans-epithelial electrical resistance (TEER) value and decreased the paracellular permeability of Caco-2 cells stimulated by lipopolysaccharide (LPS). In addition, B. dentium N8 significantly increased ZO-1, occludin, and claudin-1 mRNA expression. B. dentium N8 downregulated the mRNA expression level of TLR4 and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Furthermore, B. dentium N8 had a better protective effect on the intestinal barrier than that of E7. Comparative genomics of B. dentium N8 and E7 showed B. dentium N8 had the specific genes encoding for adhesion ability and immune system regulation. The findings provide the theoretical basis for B. dentium N8 possessing a protective effect on the intestinal barrier, which indicate that it could be used as a novel therapy for IBD.
Collapse
Affiliation(s)
- Li Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Guo W, Xiang Q, Mao B, Tang X, Cui S, Li X, Zhao J, Zhang H, Chen W. Protective Effects of Microbiome-Derived Inosine on Lipopolysaccharide-Induced Acute Liver Damage and Inflammation in Mice via Mediating the TLR4/NF-κB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7619-7628. [PMID: 34156842 DOI: 10.1021/acs.jafc.1c01781] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This research assessed the anti-inflammatory and hepatoprotective properties of inosine and the associated mechanism. Inosine pretreatment significantly reduced the secretion of several inflammatory factors and serum alanine transaminase (ALT) and aspartate amino transferase (AST) levels in a dose-dependent manner compared with the lipopolysaccharide (LPS) group. In LPS-treated mice, inosine pretreatment significantly reduced the ALT and malondialdehyde (MDA) concentration and significantly elevated the antioxidant enzyme activity. Furthermore, inosine pretreatment significantly altered the relative abundance of the genera, Bifidobacterium, Lachnospiraceae UCG-006, and Muribaculum. Correlation analysis showed that Bifidobacterium and Lachnospiraceae UCG-006 were positively related to the cecal short-chain fatty acids but negatively related to the serum IL-6 and hepatic AST and ALT levels. Notably, inosine pretreatment significantly modulated the hepatic TLR4, MYD88, NF-κB, iNOS, COX2, AMPK, Nfr2, and IκB-α expression. These results suggested that inosine pretreatment alters the intestinal microbiota structure and improves LPS-induced acute liver damage and inflammation through modulating the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Weiling Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qunran Xiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
27
|
Iridoids with anti-inflammatory effect from the aerial parts of Morinda officinalis How. Fitoterapia 2021; 153:104991. [PMID: 34265404 DOI: 10.1016/j.fitote.2021.104991] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022]
Abstract
Morinda officinalis How was widely applied to alleviate symptom like impotence, menstrual disorders, osteoporosis, and rheumatoid arthritis. To expand resources usage, phytochemistry of the aerial parts was studied and the structures of compounds were elucidated based on NMR, HRESIMS, IR and UV. Moreover, the anti-inflammatory effect and possible mechanism were investigated by Griess kit, RT-qPCR, ELISA, western blot and molecular docking on LPS-induced inflammation in RAW 264.7 cells. Herein, we isolated and identified 16 iridoid derivatives, including seven new iridoids officinaloside A-G (1-7) and nine known iridoids. All the compounds were safe to RAW 264.7 cells. Luckily, compounds 5 and 6 showed inhibitory effect on production of NO, and decreased the expression of inflammatory cytokines at mRNA and protein levels in a dose-dependent way. The possible mechanism of their anti-inflammation may be the affinity interaction between 5 with COX-2 protein, and 6 with iNOS protein. Overall, compounds 5 and 6 exert promising effects in inhibiting inflammatory cytokines, indicating that they could be used as lead compounds for developing health products or clinical practice for inflammation, which provides a scientific basis for further sustainable development and usage of the aerial parts of Morinda officinalis How.
Collapse
|
28
|
Potential Role of Probiotics in Ameliorating Psoriasis by Modulating Gut Microbiota in Imiquimod-Induced Psoriasis-Like Mice. Nutrients 2021; 13:nu13062010. [PMID: 34207960 PMCID: PMC8230682 DOI: 10.3390/nu13062010] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/02/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is an immune-mediated systemic disease that may be treated with probiotics. In this study, probiotic strains that could or could not decrease interleukin (IL)-17 levels were applied to imiquimod (IMQ)-induced psoriasis-like mice via oral administration. Bifidobacterium adolescentis CCFM667, B. breve CCFM1078, Lacticaseibacillus paracasei CCFM1074, and Limosilactobacillus reuteri CCFM1132 ameliorated psoriasis-like pathological characteristics and suppressed the release of IL-23/T helper cell 17 (Th17) axis-related inflammatory cytokines, whereas B. animalis CCFM1148, L. paracasei CCFM1147, and L. reuteri CCFM1040 neither alleviated the pathological characteristics nor reduced the levels of inflammatory cytokines. All effective strains increased the contents of short-chain fatty acids, which were negatively correlated with the levels of inflammatory cytokines. By performing 16S rRNA gene sequencing, the diversity of gut microbiota in psoriasis-like mice was found to decrease, but all effective strains made some specific changes to the composition of gut microbiota compared to the ineffective strains. Furthermore, except for B. breve CCFM1078, all other effective strains decreased the abundance of the family Rikenellaceae, which was positively correlated with psoriasis-like pathological characteristics and was negatively correlated with propionate levels. These findings demonstrated effects of strain-specificity, and how probiotics ameliorated psoriasis and provide new possibilities for the treatment of psoriasis.
Collapse
|
29
|
Zheng C, Chen T, Lu J, Wei K, Tian H, Liu W, Xu T, Wang X, Wang S, Yang R, Yang Y, Liu Z, Wei H, Deng X. Adjuvant treatment and molecular mechanism of probiotic compounds in patients with gastric cancer after gastrectomy. Food Funct 2021; 12:6294-6308. [PMID: 34052844 DOI: 10.1039/d1fo01375k] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastrectomy is the main treatment for gastric cancer (GC) at present. Surgery improves the survival rate of patients, but the complications seriously affect the recovery and lack effective treatment measures. In the present study, probiotic compounds (4 strains; Lactobacillus plantarum MH-301 (CGMCC NO. 18618), L. rhamnosus LGG-18 (CGMCC NO. 14007), L. acidophilus and Bifidobacterium animalis subsp.lactis LPL-RH (CGMCC NO. 4599)), through clinical and animal model verification, were studied to try to find the auxiliary treatment measures after gastrectomy, and explore its potential mechanism. Clinical research results showed that probiotic compounds treatment could significantly lower postoperative inflammation, enhance immunity, resume gut microbiota composition and promote postoperative recovery. The results in rat models indicated that gastrostomy led to the aggravation of inflammation, the impairment of immunity and intestinal barrier, and the disorder of gut microbiota in vivo. Furthermore, probiotic compounds' administration could downregulate the inflammatory and permeability signaling pathways in the intestinal tissue, reduce the levels of proinflammatory factors, maintain the intestinal mucosal barrier and immune function, and recover the disorder of gut microbiota after gastrectomy in rats. Therefore, we conclude that probiotic compounds can restore gut microbiota homeostasis, reduce inflammation, maintain intestinal mucosal barrier and immunity, finally promote recovery after gastrectomy, and is expected to improve the prognosis of patients.
Collapse
Affiliation(s)
- Cihua Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Geng T, He F, Su S, Sun K, Zhao L, Zhao Y, Bao N, Pan L, Sun H. Probiotics Lactobacillus rhamnosus GG ATCC53103 and Lactobacillus plantarum JL01 induce cytokine alterations by the production of TCDA, DHA, and succinic and palmitic acids, and enhance immunity of weaned piglets. Res Vet Sci 2021; 137:56-67. [PMID: 33932824 DOI: 10.1016/j.rvsc.2021.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/02/2021] [Accepted: 04/12/2021] [Indexed: 01/20/2023]
Abstract
Probiotics, including Lactobacillus rhamnosus GG ATCC53103 and Lactobacillus plantarum JL01, can improve growth performance and immunity of piglets, and relieve weaning stress-related immune disorders such as intestinal infections and inflammation. This study aimed to evaluate the ability of co-administration of the probiotics L. rhamnosus GG ATCC53103 and L. plantarum JL01 to stimulate immune responses and improve gut health during the critical weaning period in piglets. Forty-eight weaned piglets were randomly divided into four groups, and fed daily for 28 days either without, or with the two probiotics independently, or in combination. On day 28, we analyzed the cytokine and bacterial changes in intestinal mucosa and the hepatic portal vein blood metabolites of the weaned piglets. Our results showed that combined L. rhamnosus GG ATCC53103 and L. plantarum JL01 significantly increased (p < 0.05) the growth performance and expression of IL-10 and TGF-β1 mRNAs. In contrast, this treatment significantly decreased (p < 0.05) IL-1β mRNA level in the jejunum, ileum, and cecum. Furthermore, the secretion of IL-6 in the cecum, IL-1β in the jejunum, ileum, and cecum, and TNF-α in the jejunum and ileum was significantly decreased (p < 0.05). The relative abundance of Prevotella_9 and Enterococcus in ileum and cecum was significantly increased (p < 0.05). The relative abundance of Ruminococcus_1 and Ruminococcaceae_UCG-005 in cecum was significantly decreased (p < 0.05). Prevotella_9 and Enterococcus may increase the accumulation of (4Z,7Z,10Z,13Z,16Z,19Z)-4,7,10,13,16,19-docosahexaenoic acid (DHA) and tauroursodeoxycholic acid (TCDA) in portal vein blood, while Ruminococcus_1 and Ruminococcaceae_UCG-005 may decrease the accumulation of succinic and palmitic acids. These results indicate that L. rhamnosus GG ATCC53103 and L. plantarum JL01 may regulate cytokine levels by reducing the accumulation of succinic and palmitic acids and increasing the accumulation of TCDA and DHA, thereby enhancing the immunity of weaned piglets.
Collapse
Affiliation(s)
- Tingting Geng
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Feng He
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Shuai Su
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Kecheng Sun
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Lei Zhao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Yuan Zhao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Nan Bao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Li Pan
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal PRODUCTION and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
31
|
Chen X, Wu Y, Hu Y, Zhang Y, Wang S. Lactobacillus rhamnosus GG Reduces β-conglycinin-Allergy-Induced Apoptotic Cells by Regulating Bacteroides and Bile Secretion Pathway in Intestinal Contents of BALB/c Mice. Nutrients 2020; 13:nu13010055. [PMID: 33375432 PMCID: PMC7823992 DOI: 10.3390/nu13010055] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/31/2022] Open
Abstract
Allergy can cause intestinal damage, including through cell apoptosis. In this study, intestinal cell apoptosis was first observed in the β-conglycinin (β-CG) allergy model, and the effect of Lactobacillus rhamnosus GG (LGG) on reducing apoptosis of cells in the intestine and its underlying mechanisms were further investigated. Allergic mice received oral LGG daily, and intestinal tissue apoptotic cells, gut microbiota, and metabolites were evaluated six and nine days after intervention. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) analysis revealed that LGG intervention could reduce the incidence of cell apoptosis more effectively than natural recovery (NR). The results of 16S rRNA analysis indicated that LGG intervention led to an increase in the relative abundance of Bacteroides. Metabolite analysis of intestinal contents indicated that histamine, N-acetylhistamine, N(α)-γ-glutamylhistamine, phenylalanine, tryptophan, arachidonic acid malate, and xanthine were significantly decreased, and deoxycholic acid, lithocholic acid were significantly increased after the LGG intervention on β-CG allergy; the decreases in histamine and N(α)-γ-glutamylhistamine were significant compared with those of NR. In conclusion, LGG reduces apoptosis of cells induced by β-CG allergy, which may be related to regulation of Bacteroides and the bile secretion pathway.
Collapse
Affiliation(s)
- Xiaoxu Chen
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China;
| | - Yuekun Wu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.W.); (Y.H.); (S.W.)
| | - Yaozhong Hu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.W.); (Y.H.); (S.W.)
| | - Yan Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.W.); (Y.H.); (S.W.)
- Correspondence: ; Tel.: +86-22-85358445
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.W.); (Y.H.); (S.W.)
| |
Collapse
|
32
|
Alsharairi NA. The Role of Short-Chain Fatty Acids in the Interplay between a Very Low-Calorie Ketogenic Diet and the Infant Gut Microbiota and Its Therapeutic Implications for Reducing Asthma. Int J Mol Sci 2020; 21:E9580. [PMID: 33339172 PMCID: PMC7765661 DOI: 10.3390/ijms21249580] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota is well known as playing a critical role in inflammation and asthma development. The very low-calorie ketogenic diet (VLCKD) is suggested to affect gut microbiota; however, the effects of VLCKD during pregnancy and lactation on the infant gut microbiota are unclear. The VLCKD appears to be more effective than caloric/energy restriction diets for the treatment of several diseases, such as obesity and diabetes. However, whether adherence to VLCKD affects the infant gut microbiota and the protective effects thereof on asthma remains uncertain. The exact mechanisms underlying this process, and in particular the potential role of short chain fatty acids (SCFAs), are still to be unravelled. Thus, the aim of this review is to identify the potential role of SCFAs that underlie the effects of VLCKD during pregnancy and lactation on the infant gut microbiota, and explore whether it incurs significant implications for reducing asthma.
Collapse
Affiliation(s)
- Naser A Alsharairi
- Heart, Mind & Body Research Group, Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia
| |
Collapse
|