1
|
Su Z, Ma C, Ru X, Zhang S, Wu C, Huang Y, Cen H, Yin Z, Zhang J. Effects of probiotic treatment on patients and animals with chronic obstructive pulmonary disease: a systematic review and meta-analysis of randomized control trials. Front Cell Infect Microbiol 2024; 14:1411222. [PMID: 39324058 PMCID: PMC11422383 DOI: 10.3389/fcimb.2024.1411222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
Objective In recent years, the lung-gut axis has received increasing attention. The oxidative stress and systemic hypoxia occurring in chronic obstructive pulmonary disease (COPD) are related to gut dysfunction. That suggests probiotics have a potential therapeutic role in COPD. In this study, we therefore evaluated the ameliorative effects of probiotics on COPD. Methods Searches were conducted in four electronic databases, including PubMed, Cochrane Library, the NIH clinical registry Clinical Trials. Gov and EMBASE. The data extracted was analyzed statistically in this study using StataMP17 software, with mean difference (MD) chosen as the effect size for continuous variables, and the results expressed as effect sizes and their 95% confidence intervals (CIs). Standardized Mean Difference (SMD) was used if the data units were different. Results We included three randomized, controlled, double-blind clinical trials and five randomized controlled animal studies. The results show that for lung function, probiotics improved %FEV1 in COPD patients (MD = 3.02, 95%CI: 1.10, 4.93). Additionally, in inflammation, probiotics increased IL-10 (SMD = 1.99, 95%CI: 1.02, 2.96) and decreased inflammatory markers such as TNF-α (SMD= -2.64, 95%Cl: -3.38, -1.90), IL-1β (SMD= -3.49, 95%Cl: -4.58, -2.40), and IL-6 (SMD= -6.54, 95%Cl: -8.36, -4.73) in COPD animals, while having no significant effect on C-reactive protein (MD = 0.30, 95%CI: -0.71, 1.32) in COPD patients. For lung structure, probiotics significantly reduced the degree of pulmonary collagen fibers deposition in COPD animals (SMD = -2.25, 95%CI: -3.08, -1.41). Conclusion Overall, probiotics may be an additional approach that can improve COPD. Further clinical trials are needed to evaluate the efficacy, safety, and impact factors of probiotics for COPD. Systematic Review Registration https://inplasy.com/inplasy-2023-4-0023/, identifier INPLASY202340023.
Collapse
Affiliation(s)
- Ziying Su
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxi Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaosong Ru
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sijia Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuyi Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yue Huang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huijie Cen
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zihui Yin
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianping Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Liu ZM, Zhang YN, Zhang HF, Chen XJ, Peng H, Zhang XY. Restoration of the Mucosal IgA Response by Improving CD4 + T Pyroptosis Fails to Attenuate Gut Bacterial Translocation and Organ Damage After LPS Attack. Dig Dis Sci 2024; 69:798-810. [PMID: 38334934 DOI: 10.1007/s10620-024-08278-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Currently, the mechanisms of impaired gut mucosal immunity in sepsis remain unclear. Gut immunoglobulin A (IgA) is an important defense mechanism against invasive pathogens, and CD4+ T cells regulate the IgA response. AIM We aimed to verify the hypothesis indicating that CD4+ T pyroptosis induced by lipopolysaccharide (LPS) leads to an impaired gut IgA response and subsequent bacterial translocation and organ damage. METHODS Cultured CD4+ T cells and mice were manipulated with LPS, and pyroptosis was improved by A438079 or adoptive CD4+ T cell transfer. The changes demonstrated in pyroptosis-related molecules, cytotoxicity and CD4+ T cells were examined to determine CD4+ T pyroptosis. The changes demonstrated in IgA+ B cells, AID (key enzyme for immunoglobulins) and IgA production and function were examined to evaluate the IgA response. Serum biomarkers, bacterial colonies and survival analysis were detected for bacterial translocation and organ damage. RESULTS LPS attack induced CD4+ T pyroptosis, as evidenced by increased expression of P2X7, Caspase-11 and cleaved GSDMD, which elevated cytotoxicity and decreased CD4+ T cells. Decreased CD4+ T subsets (Foxp3+ T and Tfh cells) influenced the IgA response, as evidenced by lower AID expression, which decreased IgA+ B cells and IgA production and function. A438079 or cell transfer improved the IgA response but failed to reduce the translocation of gut pathogens, damage to the liver and kidney, and mortality of mice. CONCLUSION LPS attack results in CD4+ T pyroptosis. Improvement of pyroptosis restores the mucosal IgA response but fails to ameliorate bacterial translocation and organ damage.
Collapse
Affiliation(s)
- Zi-Meng Liu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi-Nan Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510089, China
| | - Hu-Fei Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510089, China
| | - Xiao-Jun Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510089, China
| | - Hui Peng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510089, China
| | - Xu-Yu Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510089, China.
| |
Collapse
|
3
|
Iancu MA, Profir M, Roşu OA, Ionescu RF, Cretoiu SM, Gaspar BS. Revisiting the Intestinal Microbiome and Its Role in Diarrhea and Constipation. Microorganisms 2023; 11:2177. [PMID: 37764021 PMCID: PMC10538221 DOI: 10.3390/microorganisms11092177] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The gut microbiota represents a community of microorganisms (bacteria, fungi, archaea, viruses, and protozoa) that colonize the gut and are responsible for gut mucosal structural integrity and immune and metabolic homeostasis. The relationship between the gut microbiome and human health has been intensively researched in the past years. It is now widely recognized that gut microbial composition is highly responsible for the general health of the host. Among the diseases that have been linked to an altered gut microbial population are diarrheal illnesses and functional constipation. The capacity of probiotics to modulate the gut microbiome population, strengthen the intestinal barrier, and modulate the immune system together with their antioxidant properties have encouraged the research of probiotic therapy in many gastrointestinal afflictions. Dietary and lifestyle changes and the use of probiotics seem to play an important role in easing constipation and effectively alleviating diarrhea by suppressing the germs involved. This review aims to describe how probiotic bacteria and the use of specific strains could interfere and bring benefits as an associated treatment for diarrhea and constipation.
Collapse
Affiliation(s)
- Mihaela Adela Iancu
- Department of Family Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Monica Profir
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Oana Alexandra Roşu
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Ruxandra Florentina Ionescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Cardiology I, “Dr. Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Bogdan Severus Gaspar
- Surgery Clinic, Emergency Clinical Hospital, 014461 Bucharest, Romania;
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Zhurilov PA, Andriyanov PA, Tutrina AI, Razheva IV, Liskova EA, Gladkova NA, Kashina DD, Yashin IV, Blokhin AA. Characterization and comparative analysis of the Escherichia marmotae M-12 isolate from bank vole (Myodes glareolus). Sci Rep 2023; 13:13949. [PMID: 37626115 PMCID: PMC10457355 DOI: 10.1038/s41598-023-41223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023] Open
Abstract
The Escherichia marmotae is a bacterium of the Enterobacterales order, which was first isolated from the Himalayan marmot (Marmota himalayana). Recently E. marmotae has been shown to cause severe infections in humans. Wild animals were suggested to be a natural reservoir of this bacterium. The present study describes the first case of E. marmotae isolation from an apparently healthy wild bank vole (Myodes glareolus). Phenotype, as well as genotype-based techniques, were applied to characterize E. marmotae M-12 isolate. E. marmotae M-12 had the capsule-positive phenotype, high adhesion to human erythrocytes and HEp-2 cells as well as a low invasion into HEp-2 cells. E. marmotae M-12 was avirulent in mice. The phylogenomic analyses of E. marmotae showed dispersed phylogenetic structure among isolates of different origins. Virulome analysis of M-12 isolate revealed the presence of the following factors: siderophores, heme uptake systems, capsule synthesis, curli and type I fimbriae, flagella proteins, OmpA porin, etc. Comparative virulome analysis among available E. marmotae genomes revealed the presence of capsule K1 genes mostly in pathogenic isolates and OmpA porin presence among all strains. We assume that the K1 capsule and OmpA porin play a key role in the virulence of E. marmotae. Pathogenesis of the latter might be similar to extraintestinal pathogenic E. coli.
Collapse
Affiliation(s)
- Pavel A Zhurilov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia.
| | - Pavel A Andriyanov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Anastasia I Tutrina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Irina V Razheva
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Elena A Liskova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Nadezda A Gladkova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Daria D Kashina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Ivan V Yashin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Andrey A Blokhin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| |
Collapse
|
5
|
Wei B, Peng Z, Xiao M, Huang T, Zheng W, Xie M, Xiong T. Limosilactobacillus fermentum NCU003089 and Lactiplantibacillus plantarum NCU001261, two probiotics with inhibition of Escherichia coli and Cronobacter sakazakii translocation in vitro. Microb Pathog 2023:106216. [PMID: 37391100 DOI: 10.1016/j.micpath.2023.106216] [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: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
The subject of this study was to screen lactic acid bacteria (LAB) with pathogen translocation inhibition and investigate the potential inhibition mechanism of it. Pathogens colonized in the intestine could cross the intestinal barrier to access blood circulation, causing severe complications. This study aimed to screen LAB with favorable inhibitory effects on the translocation of enterinvasive Escherichia coli CMCC44305 (E. coli) and Cronobacter sakazakii CMCC45401 (C. sakazakii), which were two common intestinal opportunistic pathogens. After an elaborate screening procedure including adhesion, antibacterial, and translocation assay, Limosilactobacillus fermentum NCU003089 (L. fermentum NCU3089) and Lactiplantibacillus plantarum NCU0011261 (L. plantarum NCU1261) were found to inhibit 58.38% and 66.85% of pathogen translocation, respectively. Subsequently, LAB pre-treatment suppressed the decline in TEER of Caco-2 monolayers caused by pathogens. Meanwhile, L. fermentum NCU3089 significantly inhibited claudin-1, ZO-1, and JAM-1 degradation caused by E. coli, and L. plantarum NCU1261 markedly reduced claudin-1 degradation caused by C. sakazakii. Also, the two LAB strains significantly decreased TNF-α level. In addition, L. fermentum NCU3089 but not L. plantarum NCU1261 tolerated well in the gastrointestinal fluids, and they were both sensitive or intermediate to nine common clinical antibiotics without hemolytic activity. In short, the two LAB strains could inhibit pathogen translocation by competing for adhesion sites, secreting antibacterial substances, reducing inflammatory cytokines levels, and maintaining intestinal barrier integrity. This study provided a feasible solution to prevent pathogen infection and translocation, and the two LAB strains were safe and had potential in food and pharmaceutical applications.
Collapse
Affiliation(s)
- Benliang Wei
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Zhen Peng
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China.
| | - Muyan Xiao
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Tao Huang
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Wendi Zheng
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China
| | - Tao Xiong
- State Key Laboratory of Food Science and Technology, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi, 330047, PR China.
| |
Collapse
|
6
|
Yu JT, Cheng XY, Zhan Y, Li YM, Liu WJ. Effects of Bifido, Yijunkang, Siliankang, and Golden Bifid on loperamide induced constipation. Shijie Huaren Xiaohua Zazhi 2023; 31:361-368. [DOI: 10.11569/wcjd.v31.i9.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Constipation is a common gastrointestinal disorder and a risk factor for colorectal cancer. Probiotics are the most widely used microecological agents for the treatment of constipation in clinical practice. Common intestinal microecological drugs include Bifido, Yijunkang, Siliankang, Golden Bifid, etc. They have different composition and may have different therapeutic effects on constipation.
AIM To compare the effects of the four microecological agents in a rat model of constipation and to assess the effects of these probiotics on constipation-related indicators.
METHODS Rats were randomly divided into six groups: Control group, model group, Bifido group, Yijunkang group, Siliankang group, and Golden Bifid group, with 10 rats in each group. The body weight, fecal quantity, shape, and water content, and intestinal transit rate of rats in each group were measured. Hematoxylin-eosin staining was performed to observe the pathological changes in the intestine of rats in each group. The expression level of intestinal mucin was analyzed by Alcian blue staining.
RESULTS There were no significant changes in body weight and fecal water content of rats in each group. Compared to the model group, the quantity of feces (P < 0.01), intestinal transit rate (P < 0.05), intestinal mucosal thickness (P < 0.01), and mucin expression level (P < 0.01) were significantly increased in the Bifido group. Intestinal transit rate (P < 0.05) and mucin expression (P < 0.001) were significantly increased in the Yijunkang group. Increased intestinal transit rate and mucin level were observed in the Siliankang group (P < 0.05), and increased mucin expression was observed in the Golden Bifid group (P < 0.01).
CONCLUSION The four microecological preparations improve constipation to varying degrees, of which Bifido has the best effect. Bifido can significantly increase the quantity of feces and intestinal transit rate of constipation rats, increase the expression level of mucin, promote the secretion of intestinal mucus, improve gastrointestinal peristalsis, and relieve constipation related symptoms.
Collapse
|
7
|
Ni Y, Zhang Y, Zheng L, Rong N, Yang Y, Gong P, Yang Y, Siwu X, Zhang C, Zhu L, Fu Z. Bifidobacterium and Lactobacillus improve inflammatory bowel disease in zebrafish of different ages by regulating the intestinal mucosal barrier and microbiota. Life Sci 2023; 324:121699. [PMID: 37061125 DOI: 10.1016/j.lfs.2023.121699] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
AIMS Inflammatory bowel disease (IBD) patients are accompanied by impaired intestinal barrier integrity and gut microbiota dysbiosis. Strategies targeting the gut microbiota are potential therapies for preventing and ameliorating IBD. MAIN METHODS The potential roles of two probiotic stains, Bifidobacterium longum BL986 (BL986) and Lactobacillus casei LC122 (LC122), on intestinal mucosal barrier function and microbiota in IBD zebrafish of different ages were investigated. KEY FINDINGS BL986 and LC122 treatment promoted the development and increased the microbiota diversity in larval zebrafish. Both probiotic treatment ameliorated mortality, promoted intestinal mucus secretion, and reduced the expression of inflammatory markers, thereby improving intestinal mucosal barrier function in dextran sulfate sodium salt (DSS)-induced ulcerative colitis (UC) and 2,4,6-trinitro-benzenesulfonicacid (TNBS)-induced Crohn's disease (CD) models in zebrafish. Moreover, the composition and function of microbiota were altered in IBD zebrafish, and probiotics treatment displayed prominent microbiota features. BL986 was more potent in the DSS-induced UC model, and increased the abundance of Faecalibaculum and butyric acid levels. LC122 exerted better protection against TNBS-induced CD, and increased the abundance of Enhydrobacter and acetic acid levels. Furthermore, the effect of probiotics was stronger in larval and aged zebrafish. CONCLUSION The impact of probiotics on IBD might differ from the subtypes of IBD and the age of the zebrafish, suggesting the types of disease and age should be taken into full consideration during the practical usage of probiotics.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liujie Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Nianke Rong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuru Yang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Ping Gong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Yang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xuerong Siwu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Chenyang Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Linxi Zhu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
8
|
Oladokun S, Adewole D. The effect of Bacillus subtilis and its delivery route on hatch and growth performance, blood biochemistry, immune status, gut morphology, and microbiota of broiler chickens. Poult Sci 2023; 102:102473. [PMID: 36736137 PMCID: PMC9898455 DOI: 10.1016/j.psj.2022.102473] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/16/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
This study evaluated the effect of probiotics (Bacillus subtilis fermentation extract) and its delivery route (in-feed or in ovo) on hatch and growth performance, blood biochemistry, immune status, gut morphology, and microbiota of broiler chickens. Hatching eggs were incubated for 21 d. On d 12, viable eggs were randomly allotted to 4 groups: the noninjected, in ovo saline (S), in ovo Bacillus subtilis 1 (P1), and in ovo Bacillus subtilis 2 (P2). On d 18, S, P1, and P2 groups received 0.2 mL saline diluent, 10 × 106, and 20 × 106 CFU of the bacterium via the amnion, respectively. At hatch, chicks were re-allotted to 5 new treatment groups: P1, P2, 0.005% in-feed Bacillus subtilis extract (P3), 0.05% in-feed bacitracin methylene disalicylate (BMD,), and corn-wheat-soybean diet negative control (NC) in 9 replicate pens (22 birds/pen) and raised for 35 d. Hatch parameters were assessed on d 0, and growth performance indices measured weekly. On d 25, 1 bird/cage was euthanized, and samples collected for further analysis. Data were analyzed by generalized linear model. Treatments S and P2 recorded higher (P = 0.01) chick BW/ Egg Weight values compared to the non-injected eggs. P3 and P2 reduced (P = 0.02) FI at week 5 compared to the NC treatment. However, no change in average body weight gain (ABG) and feed conversion ratio (FCR) were observed during the same period. At d 35, while BMD treatment showed a tendency (P = 0.09) to increase FI compared to the NC treatment, ABG and FCR were similar for all treatments. Blood sodium and chloride levels were increased (P < 0.05) by the BMD treatment compared to the NC treatment. Compared to other treatments, BMD and P3 treatments increased (P < 0.001) jejunal and ileal villus height to crypt depth ratios, respectively. However, P1 and P2 increased (P < 0.001) villus height to crypt depth ratio in the duodenum compared to NC treatment. Treatments did not affect gut microbial diversity; however, BMD treatment increased (P < 0.05) the proportion of bacteria in the genus Enterococcus in the ileum and reduced (P < 0.05) the proportion of bacteria in the genus Streptococcus in the ceca. All probiotics treatments (irrespective of route and dose) reduced (P < 0.001) the levels of serum IgG compared to the NC treatment. However, P1 and P2 had the lowest numerical decrease in serum IgG concentrations, suggesting that Bacillus subtilis (especially in ovo delivered) might provide broiler chickens with better immunological protection by neutralizing pathogenic organisms that could result in the production of natural antibodies.
Collapse
Affiliation(s)
- Samson Oladokun
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS B2N 5E3, Canada
| | - Deborah Adewole
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS B2N 5E3, Canada.
| |
Collapse
|
9
|
Liu J, Shi L, Tuo X, Ma X, Hou X, Jiang S, Lv J, Cheng Y, Guo D, Han B. Preparation, characteristic and anti-inflammatory effect of selenium nanoparticle-enriched probiotic strain Enterococcus durans A8-1. J Trace Elem Med Biol 2022; 74:127056. [PMID: 35939922 DOI: 10.1016/j.jtemb.2022.127056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/01/2022] [Accepted: 08/02/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Elemental selenium, a new type of selenium supplement, can be biosynthesized via microorganisms. This study is to characterize a patent probiotic bacteria Enterococcus durans A8-1, capable of reducing selenite (Se6+ or Se4+) to elemental selenium (Se0) with the formation of Se nanoparticles (SeNPs). METHODS The selenium nanoparticles synthesized from A8-1 were characterized using scanning electron microscopy (SEM), transmission electron microscopy (TEM), energy dispersive spectroscopy (EDS), and X-ray photoelectron energy (XPS). The Caco2 cells were used to investigate the effects of Se-enriched A8-1 on the viability, membrane integrity, and the regulation of cellular inflammation through MTT and ELISA assays. The selenium-enriched metabolic function of A8-1 was analyzed by transcriptome sequencing. RESULTS E. durans A8-1 has the ability to synthesize intracellular SeNPs that are incubated with 60 mg/L sodium selenite for 18 h at 37 °C with 7 % inoculum under aerobic conditions. The selenium-enriched transformation rate increased to 43.46 %. After selenium enrichment, there were no significant morphological changes in E. durans A8-1 cells. The cells also exhibited no cytotoxicity when incubated with Caco-2 cells, and increased cellular proliferation. Furthermore, Se-enriched A8-1 cells antagonize the adhesion of S. typhimurium ATCC14028 onto the surface of Caco-2 cells protecting cell membrane integrity and was assessed by measuring LDH and AKP activities (P <0.001, P <0.001). Moreover, Se-enriched A8-1 could protect Caco-2 cells from inflammation induced by lipopolysaccharide and help the cells alleviate the inflammation through the reduced expression of cytokine IL-8 (P = 0.0012, P <0.001) and TNF-α (P <0.001, P <0.001). Based on transcriptome sequencing in Se-enriched E. durans A8-1 cells, there were 485 up-regulated genes and 322 down-regulated genes (Padj < 0.05). There were 19 predicted up-regulated genes that are highly related to the potential selenium metabolism pathway, which focuses on the transportation of Na2SeO3 by membrane proteins, and gradually reduces Na2SeO3 to elemental selenium aggregates that are deposited onto the membrane surface via the intracellular redox response. CONCLUSION E. durans A8-1 could convert extracellular selenite into intracellular biological SeNPs via redox pathway with strong selenium-rich metabolism, and its biological SeNPs have anti-inflammatory properties, which have the potential for the development of composite selenium nanomaterials and can be further studied for the function of SeNPs with potential applications.
Collapse
Affiliation(s)
- Jin Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lu Shi
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaohong Tuo
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xinxin Ma
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xinyao Hou
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sijin Jiang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jia Lv
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yue Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China
| | - Dagang Guo
- School of Material Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Bei Han
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China.
| |
Collapse
|
10
|
Ye T, Yuan S, Kong Y, Yang H, Wei H, Zhang Y, Jin H, Yu Q, Liu J, Chen S, Sun J. Effect of Probiotic Fungi against Cognitive Impairment in Mice via Regulation of the Fungal Microbiota-Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9026-9038. [PMID: 35833673 DOI: 10.1021/acs.jafc.2c03142] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The fungal microbiota may be involved in the regulation of cognition and behavior, while the role of probiotic fungi against cognitive impairment is unclear. Here, we explored the idea that probiotic Saccharomyces boulardii could participate in the regulation of microglia-induced neuroinflammation in Alzheimer's disease (AD) model mice. Cognitive deficits, deposits of amyloid-β (Aβ) and phosphorylation of tau, synaptic plasticity, microglia activation, and neuroinflammatory reactions were observed. The expression levels of Toll-like receptors (TLRs) pathway-related proteins were detected. Meanwhile, intestinal barrier integrity and fungal microbiota composition were evaluated. Our results showed fungal microbiota dysbiosis in APP/PS1 mice, which might result in the neuroinflammation of AD. The increased levels of interleukin (IL)-6, IL-1β, and cluster of differentiation 11b (CD11b) were observed in APP/PS1 mice, which were associated with activation of microglia, indicative of a broader recognition of neuroinflammation mediated by fungal microbiota compared to hitherto appreciated. Probiotic S. boulardii treatment improved dysbiosis, alleviated the neuroinflammation as well as synaptic injury, and ultimately improved cognitive impairment. Moreover, S. boulardii therapy could inhibit microglia activation and the TLRs pathway, which were reversed by antifungal treatment. These findings revealed that S. boulardii actively participated in regulating the TLRs pathway to inhibit the neuroinflammation via the gut-brain axis.
Collapse
Affiliation(s)
- Tao Ye
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shushu Yuan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu Kong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huiqun Yang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongming Wei
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yuhe Zhang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hangqi Jin
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qingxia Yu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Songfang Chen
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jing Sun
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
11
|
Yang Z, Liu X, Wu Y, Peng J, Wei H. Effect of the Microbiome on Intestinal Innate Immune Development in Early Life and the Potential Strategy of Early Intervention. Front Immunol 2022; 13:936300. [PMID: 35928828 PMCID: PMC9344006 DOI: 10.3389/fimmu.2022.936300] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
Early life is a vital period for mammals to be colonized with the microbiome, which profoundly influences the development of the intestinal immune function. For neonates to resist pathogen infection and avoid gastrointestinal illness, the intestinal innate immune system is critical. Thus, this review summarizes the development of the intestinal microbiome and the intestinal innate immune barrier, including the intestinal epithelium and immune cells from the fetal to the weaning period. Moreover, the impact of the intestinal microbiome on innate immune development and the two main way of early-life intervention including probiotics and fecal microbiota transplantation (FMT) also are discussed in this review. We hope to highlight the crosstalk between early microbial colonization and intestinal innate immunity development and offer some information for early intervention.
Collapse
Affiliation(s)
- Zhipeng Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangchen Liu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yanting Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
12
|
Zhu Y, Xu Y, Wang X, Rao L, Yan X, Gao R, Shen T, Zhou Y, Kong C, Zhou L. Probiotic Cocktail Alleviates Intestinal Inflammation Through Improving Gut Microbiota and Metabolites in Colitis Mice. Front Cell Infect Microbiol 2022; 12:886061. [PMID: 35782138 PMCID: PMC9240319 DOI: 10.3389/fcimb.2022.886061] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
The modulation of the gut microbiome has been widely suggested as a promising therapeutic strategy for inflammatory bowel disease (IBD). Here, we established a novel probiotic cocktail to investigate its therapeutic role in acute colitis mice. During dextran sulfate sodium (DSS)-induced colitis, the mice were treated with the probiotic cocktail, fecal microbiota transplantation (FMT) from a healthy mice donor, or 5-aminosalicylic acid (5-ASA), respectively. The inflammatory responses were assessed by symptoms, serum inflammatory factors, and histological scoring. The intestinal barrier function was assessed by detecting tight junction proteins. Gut microbiota and its metabolites were further identified using 16S rDNA sequencing and a liquid chromatograph mass spectrometer (LC-MS/MS). Compared with FMT and 5-ASA treatment, the probiotic cocktail performed better in alleviating symptoms of colitis and decreasing disease activity score and mucosal inflammation. The probiotic cocktail also significantly decreased serum IL-17 level and increased JAM-1 expression in colon. The gut microbiota analysis confirmed that the beneficial effects of the probiotic cocktail were attributed to increasing anti-inflammatory bacteria Akkermansia, Bifidobacterium, and Blautia, while decreasing pro-inflammatory bacteria Parasutterella. The targeted metabolome analysis further indicated a rise in the production of Bifidobacterium-related short-chain fatty acids (SCFAs) such as propanoic acid and isobutyric acid after probiotics treatment. Taken together, the probiotic cocktail effectively alleviated intestinal inflammation through improving gut microbiota and metabolites in colitis mice, suggesting its great potential to be a novel therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- Yefei Zhu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Yu Xu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Xinyue Wang
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Leiping Rao
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xuebing Yan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Renyuan Gao
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Tongyi Shen
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Yuan Zhou
- School of Biological Engineering, Hefei Technology College, Hefei, China
| | - Cheng Kong
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| | - Longxiang Zhou
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| |
Collapse
|
13
|
Khalifa A, Sheikh A, Ibrahim HIM. Bacillus amyloliquefaciens Enriched Camel Milk Attenuated Colitis Symptoms in Mice Model. Nutrients 2022; 14:1967. [PMID: 35565934 PMCID: PMC9101272 DOI: 10.3390/nu14091967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/30/2022] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
Fermented camel's milk has various health beneficial prebiotics and probiotics. This study aimed to evaluate the preventive efficacy of Bacillus amyloliquefaciens enriched camel milk (BEY) in 2-, 4- and 6-Trinitrobenzenesulfonic acid (TNBS)-induced colitis mice models. To this end, the immune modulatory effects of Bacillus amyloliquefaciens (BA) on TNF-α challenged HT29 colon cells were estimated using the cell proliferation and cytokines ELISA method. BEY was prepared using the incubation method and nutritional value was quantified by comparing it to commercial yogurt. Furthermore, TNBS-induced colitis was established and the level of disease index, pathological scores, and inflammatory markers of BEY-treated mice using macroscopic and microscopic examinations, qPCR and immunoblot were investigated. The results demonstrate that BA is non-toxic to HT29 colon cells and balanced the inflammatory cytokines. BEY reduced the colitis disease index, and improved the body weight and colon length of the TNBS-induced mice. Additionally, Myeloperoxidase (MPO) and pro-inflammatory cytokines (IL1β, IL6, IL8 and TNF-α) were attenuated by BEY treatment. Moreover, the inflammatory progress mRNA and protein markers nuclear factor kappa B (NFκB), phosphatase and tensin homolog (PTEN), proliferating cell nuclear antigen (PCNA), cyclooxygenase-2 (COX-2) and occludin were significantly down-regulated by BEY treatment. Interestingly, significant suppression of PCNA was observed in colonic tissues using the immunohistochemical examination. Treatment with BEY increased the epigenetic (microRNA217) interactions with PCNA. In conclusion, the BEY clearly alleviated the colitis symptoms and in the future could be used to formulate a probiotic-based diet for the host gut health and control the inflammatory bowel syndrome in mammals.
Collapse
Affiliation(s)
- Ashraf Khalifa
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Abdullah Sheikh
- Camel Research Center, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
| | - Hairul Islam Mohamed Ibrahim
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Molecular Biology Division, Pondicherry Centre for Biological Sciences and Educational Trust, Kottakuppam 605104, India
| |
Collapse
|
14
|
He J, Guo K, Chen Q, Wang Y, Jirimutu. Camel milk modulates the gut microbiota and has anti-inflammatory effects in a mouse model of colitis. J Dairy Sci 2022; 105:3782-3793. [PMID: 35248376 DOI: 10.3168/jds.2021-21345] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/13/2022] [Indexed: 01/03/2024]
Abstract
Camel milk is a nutritionally rich food that shows anti-inflammatory, immune regulation, and gut microbiota maintenance properties. However, the relationship between camel milk and the intestinal microbiota during colitis is unclear. Herein, we evaluated the protective effect of camel milk in mice with colitis induced using dextran sodium sulfate. Our results showed that camel milk can prevent body weight loss and colon shortening, reduce the disease activity index, and attenuate colon tissue damage. Additionally, camel milk could reduce the overexpression of inflammatory factors, inhibit the apoptosis of intestinal epithelial cells, and promote the expression of claudin-1, occludin, and zonula occludens-1 proteins. Moreover, camel milk effectively regulated intestinal microbiota in mice with colitis by increasing the gut microbiota diversity, increasing the abundance of beneficial bacteria (such as g_norank_f_Muribaculaceae, and Lachnospiraceae_NK4A136_group), and reducing the number of harmful bacteria (Bacteroides, Escherichia-Shigella). In addition, camel milk increased the levels of intestinal short-chain fatty acids. The results of the present study demonstrated that via regulating the intestinal microbiota, maintaining intestinal barrier function, and inhibiting proinflammatory cytokines, camel milk can ameliorate dextran sodium sulfate-induced colitis.
Collapse
Affiliation(s)
- Jing He
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010010, China
| | - Kunjie Guo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010010, China
| | - Qi Chen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010010, China
| | - Yulin Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010010, China
| | - Jirimutu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010010, China; Camel Research Institute of Inner Mongolia, Alashan, Inner Mongolia, 750330, China.
| |
Collapse
|
15
|
Jin S, Wetzel D, Schirmer M. Deciphering mechanisms and implications of bacterial translocation in human health and disease. Curr Opin Microbiol 2022; 67:102147. [PMID: 35461008 DOI: 10.1016/j.mib.2022.102147] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022]
Abstract
Significant increases in potential microbial translocation, especially along the oral-gut axis, have been identified in many immune-related and inflammatory diseases, such as inflammatory bowel disease, colorectal cancer, rheumatoid arthritis, and liver cirrhosis, for which we currently have no cure or long-term treatment options. Recent advances in computational and experimental omics approaches now enable strain tracking, functional profiling, and strain isolation in unprecedented detail, which has the potential to elucidate the causes and consequences of microbial translocation. In this review, we discuss current evidence for the detection of bacterial translocation, examine different translocation axes with a primary focus on the oral-gut axis, and outline currently known translocation mechanisms and how they adversely affect the host in disease. Finally, we conclude with an overview of state-of-the-art computational and experimental tools for strain tracking and highlight the required next steps to elucidate the role of bacterial translocation in human health.
Collapse
Affiliation(s)
- Shen Jin
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Daniela Wetzel
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Melanie Schirmer
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
16
|
Zhou A, Yuan Y, Yang M, Huang Y, Li X, Li S, Yang S, Tang B. Crosstalk Between the Gut Microbiota and Epithelial Cells Under Physiological and Infectious Conditions. Front Cell Infect Microbiol 2022; 12:832672. [PMID: 35155283 PMCID: PMC8829037 DOI: 10.3389/fcimb.2022.832672] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract (GIT) is considered the largest immunological organ, with a diverse gut microbiota, that contributes to combatting pathogens and maintaining human health. Under physiological conditions, the crosstalk between gut microbiota and intestinal epithelial cells (IECs) plays a crucial role in GIT homeostasis. Gut microbiota and derived metabolites can compromise gut barrier integrity by activating some signaling pathways in IECs. Conversely, IECs can separate the gut microbiota from the host immune cells to avoid an excessive immune response and regulate the composition of the gut microbiota by providing an alternative energy source and releasing some molecules, such as hormones and mucus. Infections by various pathogens, such as bacteria, viruses, and parasites, can disturb the diversity of the gut microbiota and influence the structure and metabolism of IECs. However, the interaction between gut microbiota and IECs during infection is still not clear. In this review, we will focus on the existing evidence to elucidate the crosstalk between gut microbiota and IECs during infection and discuss some potential therapeutic methods, including probiotics, fecal microbiota transplantation (FMT), and dietary fiber. Understanding the role of crosstalk during infection may help us to establish novel strategies for prevention and treatment in patients with infectious diseases, such as C. difficile infection, HIV, and COVID-19.
Collapse
Affiliation(s)
- An Zhou
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Yuan
- Institution of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Min Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yujiao Huang
- The First Clinical College, ChongQing Medical University, Chongqing, China
| | - Xin Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shengpeng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- *Correspondence: Shiming Yang, ; Bo Tang,
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- *Correspondence: Shiming Yang, ; Bo Tang,
| |
Collapse
|
17
|
Supplemental Bacillus subtilis PB6 Improves Growth Performance and Gut Health in Broilers Challenged with Clostridium perfringens. J Immunol Res 2021; 2021:2549541. [PMID: 34746321 PMCID: PMC8566084 DOI: 10.1155/2021/2549541] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
Clostridium perfringens (CP) is the principal pathogenic bacterium of chicken necrotic enteritis (NE), which causes substantial economic losses in poultry worldwide. Although probiotics are known to provide multiple benefits, little is known about the potential effects of Bacillus subtilis (B. subtilis) application in preventing CP-induced necrotic enteritis. In this study, 450 male Arbor Acres broilers were divided into 5 experimental treatments: A: basal diet (control group); B: basal diet and CP challenge (model group); C: CP challenge+10 mg/kg enramycin (positive control group); D: CP challenge+4 × 107 CFU/kg of feed B. subtilis PB6 (PB6 low-dosage group); and E: CP challenge+6 × 107 CFU/kg of feed B. subtilis PB6 (PB6 high-dosage group). There were 6 replicate pens per treatment with 15 broilers per pen. The present research examined the effect of Bacillus subtilis PB6 (B. subtilis PB6) on growth performance, mRNA expression of intestinal cytokines and tight junctions, and gut flora composition in broilers challenged with CP. The entire experiment was divided into two phases: the non-CP challenge phase (d0–18) and the CP challenge phase (d18–26). PB6 did not increase the growth performance during the first stage, but the PB6 high-dosage group was found to have larger body weight gain and ADFI during the CP challenge stage. Feed supplementation with PB6 reduced the lesion score of challenged chicks, with increased tight junction-related gene expression (occludin and ZO-1) and decreased TNF-α expression compared with CP-infected birds. A decrease in the abundance of Clostridium XI, Streptococcus, and Staphylococcus was observed after CP infection (P < 0.05), while supplementation with PB6 restored the ileal microbial composition. In conclusion, administration of B. subtilis PB6 improved growth performance, enhanced intestinal barrier function, and mitigated intestinal inflammation/lesions, which might be due to its restoring effects on the ileal microbial composition in CP-challenged broilers.
Collapse
|
18
|
Chen B, Luo J, Han Y, Du H, Liu J, He W, Zhu J, Xiao J, Wang J, Cao Y, Xiao H, Song M. Dietary Tangeretin Alleviated Dextran Sulfate Sodium-Induced Colitis in Mice via Inhibiting Inflammatory Response, Restoring Intestinal Barrier Function, and Modulating Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7663-7674. [PMID: 34182753 DOI: 10.1021/acs.jafc.1c03046] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this study, the preventive effect of tangeretin (TAN), a natural flavonoid derivative from citrus fruits, on the dextran sulfate sodium (DSS)-induced colitis in mice was evaluated. Our results showed that dietary TAN (0.04% and 0.08% w/w in the diet) significantly reduced the severity of colitis caused by DSS treatment in mice, evidenced by the increased colon length, the reduced disease activity index, and the attenuated colonic tissue damages. Moreover, dietary TAN inhibited the inflammatory response via down-regulating the overexpression of colonic inflammatory cytokines. Immunohistochemical analysis revealed that the intestinal barrier function was restored by TAN through enhancing claudin-1 and ZO-1 expressions. Additionally, dietary TAN modulated gut microbiota in colitic mice via enhancing gut microbiota diversity, ascending the level of beneficial bacteria, e.g., Lachnospiraceae and Lactobacillaceae, and descending the level of harmful bacteria, e.g., Enterobacteriaceae and Alistipes. Besides, dietary TAN promoted short-chain fatty acids production in DSS-treated mice. Altogether, these findings provided scientific evidence for the rational utilization of TAN as a preventive agent against colonic inflammation and related diseases.
Collapse
Affiliation(s)
- Bin Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Jiakang Luo
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jie Liu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University (BTBU), Beijing 100048, P. R. China
| | - Wei He
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Jinhui Zhu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University (BTBU), Beijing 100048, P. R. China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, P.R. China
| |
Collapse
|
19
|
Li Y, Xia S, Jiang X, Feng C, Gong S, Ma J, Fang Z, Yin J, Yin Y. Gut Microbiota and Diarrhea: An Updated Review. Front Cell Infect Microbiol 2021; 11:625210. [PMID: 33937093 PMCID: PMC8082445 DOI: 10.3389/fcimb.2021.625210] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Diarrhea is a common problem to the whole world and the occurrence of diarrhea is highly associated with gut microbiota, such as bacteria, fungi, and viruses. Generally, diarrheal patients or animals are characterized by gut microbiota dysbiosis and pathogen infections may lead to diarrheal phenotypes. Of relevance, reprograming gut microbiota communities by dietary probiotics or fecal bacteria transplantation are widely introduced to treat or prevent diarrhea. In this review, we discussed the influence of the gut microbiota in the infection of diarrhea pathogens, and updated the research of reshaping the gut microbiota to prevent or treat diarrhea for the past few years. Together, gut microbiota manipulation is of great significance to the prevention and treatment of diarrhea, and further insight into the function of the gut microbiota will help to discover more anti-diarrhea probiotics.
Collapse
Affiliation(s)
- Yunxia Li
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Siting Xia
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaohan Jiang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Can Feng
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Saiming Gong
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jie Ma
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Jie Yin, ; Zhengfeng Fang,
| | - Jie Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- *Correspondence: Jie Yin, ; Zhengfeng Fang,
| | - Yulong Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
20
|
Kunyeit L, K A AA, Rao RP. Application of Probiotic Yeasts on Candida Species Associated Infection. J Fungi (Basel) 2020; 6:jof6040189. [PMID: 32992993 PMCID: PMC7711718 DOI: 10.3390/jof6040189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023] Open
Abstract
Superficial and life-threatening invasive Candida infections are a major clinical challenge in hospitalized and immuno-compromised patients. Emerging drug-resistance among Candida species is exacerbated by the limited availability of antifungals and their associated side-effects. In the current review, we discuss the application of probiotic yeasts as a potential alternative/ combination therapy against Candida infections. Preclinical studies have identified several probiotic yeasts that effectively inhibit virulence of Candida species, including Candida albicans, Candida tropicalis, Candida glabrata, Candida parapsilosis, Candida krusei and Candida auris. However, Saccharomyces cerevisiae var. boulardii is the only probiotic yeast commercially available. In addition, clinical studies have further confirmed the in vitro and in vivo activity of the probiotic yeasts against Candida species. Probiotics use a variety of protective mechanisms, including posing a physical barrier, the ability to aggregate pathogens and render them avirulent. Secreted metabolites such as short-chain fatty acids effectively inhibit the adhesion and morphological transition of Candida species. Overall, the probiotic yeasts could be a promising effective alternative or combination therapy for Candida infections. Additional studies would bolster the application of probiotic yeasts.
Collapse
Affiliation(s)
- Lohith Kunyeit
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Anu-Appaiah K A
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Reeta P. Rao
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
- Correspondence: ; Tel.: +1-508-831-5000
| |
Collapse
|
21
|
Peng Z, Wei B, Huang T, Liu Z, Guan Q, Xie M, Li H, Xiong T. Screening, Safety Evaluation, and Mechanism of Two Lactobacillus fermentum Strains in Reducing the Translocation of Staphylococcus aureus in the Caco-2 Monolayer Model. Front Microbiol 2020; 11:566473. [PMID: 33042071 PMCID: PMC7524899 DOI: 10.3389/fmicb.2020.566473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/14/2020] [Indexed: 01/05/2023] Open
Abstract
Staphylococcus aureus is a common commensal of humans, and its translocation from gastrointestine to peripheral organs and tissues could cause severe diseases and complications. This study focuses on the screening and characterization of Lactobacillus strains with significant inhibitory effect on the translocation of S. aureus through Caco-2 monolayers. First, strains with strong affinity for mucin and Caco-2 cells were obtained, via microtiter plate assay and adhesion assay, respectively. Obtained bacteria were further tested for their inhibitory effects on the growth of S. aureus by well diffusion assay. Subsequently, two strains preincubated with Caco-2 monolayers were found to inhibit the translocation of S. aureus CMCC26003 by 80.95 and 43.96%, respectively, via the transcellular translocation assay. These two strains were then identified to be Lactobacillus fermentum NCU3087 and L. fermentum NCU3088. Second, the mechanism of inhibition was investigated by analyzing the relative concentration of tight junction proteins and proinflammatory cytokines of Caco-2 cells, by Western blot and enzyme-linked immunosorbent assay, respectively. Results showed that both NCU3087 and NCU3088 significantly attenuated the degradation of occludin, claudin-1, ZO-1, and JAM-1 and suppressed the secretion of interleukin 6 and tumor necrosis factor-α induced by S. aureus, to different extent. Moreover, two Lactobacillus strains could barely translocate the Caco-2 monolayers, had no hemolytic activity, displayed strong resistance to gastrointestinal fluids, and were sensitive or moderate sensitive to nine clinically relevant antibiotics. Collectively, this study identified two Lactobacillus strains with significant inhibitory effect on the translocation of S. aureus, and their safeness for application was evaluated, thereby providing potential solutions for prevention of S. aureus and prophylaxis of related diseases.
Collapse
Affiliation(s)
- Zhen Peng
- School of Food Science and Technology, Nanchang University, Nanchang, China.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Benliang Wei
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Tao Huang
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhanggen Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Qianqian Guan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Mingyong Xie
- School of Food Science and Technology, Nanchang University, Nanchang, China.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Haijuan Li
- College of Biological and Environmental Engineering, Xi'an University, Xi'an, China
| | - Tao Xiong
- School of Food Science and Technology, Nanchang University, Nanchang, China.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Gong H, Yuan Q, Pang J, Li T, Li J, Zhan B, Chang R, Mao X. Dietary Milk Fat Globule Membrane Restores Decreased Intestinal Mucosal Barrier Development and Alterations of Intestinal Flora in Infant-Formula-Fed Rat Pups. Mol Nutr Food Res 2020; 64:e2000232. [PMID: 32918844 DOI: 10.1002/mnfr.202000232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SCOPE Milk fat globule membrane (MFGM), which contains abundant polar lipids and glycoproteins, can narrow the gap in growth and development between breast-fed and infant-formula-fed babies. The objective of this study is to evaluate the effect of MFGM supplementation in infant formula on intestinal epithelium maturation, tight junctions, and gut colonization in rat pups. METHODS AND RESULTS Sprague Dawley rat pups consume one of the five diets from postnatal day 8, including rat breastfeeding (BF), infant formula (IF), and infant formula containing MFGM at 260 mg kg-1 body weight (BW), 520 mg kg-1 BW, or 1040 mg kg-1 BW. Results show that MFGM supplementation in infant formula can facilitate intestinal mucosal barrier maturation via promoting intestinal proliferation and differentiation, and increasing tight junction proteins. In addition, compared with that of the IF pups, the intestinal flora composition of MFGM-supplemented pups is more similar to that of BF pups. CONCLUSION MFGM supplementation in infant formula can restore the intestinal development in infant-formula-fed pups, which suggests that the supplementation of MFGM in infant formula can better mimic breast milk.
Collapse
Affiliation(s)
- Han Gong
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| | - Qichen Yuan
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| | - Jinzhu Pang
- Mengniu Arla (Inner Mongolia) Dairy Products Co., Ltd., Beijing, 101100, China
| | - Tiange Li
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| | - Jufang Li
- Mengniu Arla (Inner Mongolia) Dairy Products Co., Ltd., Beijing, 101100, China
| | - Biyuan Zhan
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| | - Rui Chang
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| | - Xueying Mao
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, China Agricultural University, Beijing, 100083, China
| |
Collapse
|
23
|
Fu CY, Li LQ, Yang T, She X, Ai Q, Wang ZL. Autoinducer-2 May Be a New Biomarker for Monitoring Neonatal Necrotizing Enterocolitis. Front Cell Infect Microbiol 2020; 10:140. [PMID: 32373545 PMCID: PMC7179697 DOI: 10.3389/fcimb.2020.00140] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Autoinducer-2 (AI-2) has a widely accepted role in bacterial intra- and interspecies communication. Little is known about the relationships between AI-2 and NEC. This study found that AI-2 levels in patients and in a NEC mouse model were detected using the Vibrio harveyi BB170 assay system. Bacterial communities of the newborns' stool microbiota (NEC acute group, NEC recovery group, control group, and antibiotics-free group) and of the NEC mouse model (NEC group and control group) were detected by high-throughput sequencing. Intestinal histopathological changes were observed after HE staining. The AI-2 level in the NEC acute group (44.75 [40.17~65.52]) was significantly lower than that in the control group, NEC recovery group and antibiotics-free group. The overall microbiota compositions of each group at the phylum level were not significantly different. The proportions of Enterococcus, Clostridium_sensu_stricto_1, Peptoclostridium, and Veillonella had significant differences among the 4 groups at the genus level. In animal experiments, the AI-2 level in feces of NEC mice (56.89 ± 11.87) was significantly lower than that in the feces of control group mice (102.70 ± 22.97). The microbiota compositions of NEC and control group mice at the phylum level were not significantly different. At the genus level, Klebsiella, Clostridium_sensu_stricto_1, and Peptoclostridium abundances in the NEC group increased significantly compared with those in the control group (P < 0.05). In addition, Lactobacillus, Pasteurella, and Parabacteroides abundances in the NEC group decreased significantly compared with those in the normal control group (P < 0.05), while Lactobacillus, Pasteurella, and Parabacteroides abundances had the opposite trend. The AI-2 concentration decreased significantly in the acute phase of NEC and increased gradually in the convalescent phase. We conclude that the concentration of AI-2 was correlated with intestinal flora disorder and different stages of disease. AI-2 may be a new biomarker for the diagnosis and monitoring of NEC. Trial Registry: ClinicalTrials.gov; ChiCTR-ROC-17013746; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Chun-Yan Fu
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Lu-Quan Li
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Ting Yang
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Xiang She
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Qing Ai
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Zheng-Li Wang
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| |
Collapse
|
24
|
张 汉, 高 杰, 何 肖, 龚 泽, 万 宇, 胡 彤, 李 煜, 曹 虹. [The postbiotic HM0539 from Lactobacillus rhamnosus GG prevents intestinal infection by enterohemorrhagic E. coli O157: H7 in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:211-218. [PMID: 32376527 PMCID: PMC7086141 DOI: 10.12122/j.issn.1673-4254.2020.02.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the protective effect of the novel postbiotic HM0539 from Lactobacillus rhamnosus GG against intestinal infection by enterohemorrhagic E. coli O157: H7. METHODS We performed adhesion and invasion experiments to evaluate whether HM0539 could block E. coli O157: H7 adhesion to HT-29 cells. The expressions of mucin2 and the tight junction proteins ZO-1 and Occludin in HM0539-treated HT-29 cells were analyzed using immunofluorescence assay and Western blotting. Animal experiments were conducted in mice to observe the survival rate and changes in body weight, intestinal morphology and the intestinal barrier function after the challenge and HM0539 treatment. RESULTS HM0539 significantly inhibited the adhesion and invasion of E. coli O157: H7 to HT-29 cells in a dose-dependent manner. HM0539 treatment 4 h prior to E. coli O157: H7 challenge significantly lowered the adhesion and invasion rates of bacteria as compared with the treatment administered at the same time of challenge (P < 0.05). E. coli O157: H7-induced down-regulation of mucin2 and tight junction proteins in HT-29 cells was obviously alleviated by HM0539 treatment of (P < 0.05). In the animal experiment, HM0539 treatment significantly inhibited body weight loss (P < 0.05), alleviated jejunal injury, and inhibited E. coli O157: H7-induced destruction of jejunal goblet cells in the challenged mice (P < 0.05). HM0539 also significantly up-regulated the expression of mucin2 and ZO-1 proteins in the jejunum of E. coli O157:H7-infected mice (P < 0.05). CONCLUSIONS HM0539 not only inhibits the adhesion and invasion of E. coli O157: H7 to HT-29 cells, but also enhances the resistance against E. coli O157: H7 infection in mice by attenuating the destruction of mucin and tight junction proteins.
Collapse
Affiliation(s)
- 汉运 张
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 杰 高
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 肖龙 何
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 泽龙 龚
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 宇 万
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 彤彤 胡
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 煜彬 李
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - 虹 曹
- />南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
25
|
Zhang S, Hu H, He W, Muhammad Z, Wang L, Liu F, Pan S. Regulatory Roles of Pectin Oligosaccharides on Immunoglobulin Production in Healthy Mice Mediated by Gut Microbiota. Mol Nutr Food Res 2019; 63:e1801363. [PMID: 31116489 DOI: 10.1002/mnfr.201801363] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/27/2019] [Indexed: 12/11/2022]
Abstract
SCOPE The prebiotic regulation of the gut microbiota is a promising strategy to induce protective humoral and mucosal immune responses. The potential immune-improving effects of pectin oligosaccharides (POS) in healthy mice and the potential mechanism mediated by specific intestinal bacteria are investigated. METHODS AND RESULTS POS is prepared using a hydrogen-peroxide-assisted degradation. Mice that consumed diets containing POS are tested for microbial community shifts, short-chain fatty acids (SCFAs), and immunoglobulin (Ig) production using quantitative real-time polymerase chain reaction, gas chromatography, and ELISA kits. Pearson's correlation analyses are performed between Ig production and specific intestinal bacteria or SCFAs. POS treatment significantly improves the growth of healthy mice. Moreover, 4-week POS administration results in a profound change in intestinal microbial composition and a significantly higher fecal concentration of acetate, which leads to substantial increases of the levels of fecal secretory immunoglobulin A and serum IgG. CONCLUSIONS The results suggest that the inclusion of POS in a diet can increase Ig production and optimize the composition of the gut microbiota. A significant correlation is observed between changes in Ig production and specific intestinal bacteria or acetate, providing insight into the mechanism of POS as a potential immune-enhancing supplement.
Collapse
Affiliation(s)
- Shanshan Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Haijuan Hu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Wanying He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Zafarullah Muhammad
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Lufeng Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Fengxia Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| | - Siyi Pan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P. R. China
| |
Collapse
|
26
|
Yang RC, Qu XY, Xiao SY, Li L, Xu BJ, Fu JY, Lv YJ, Amjad N, Tan C, Kim KS, Chen HC, Wang XR. Meningitic Escherichia coli-induced upregulation of PDGF-B and ICAM-1 aggravates blood-brain barrier disruption and neuroinflammatory response. J Neuroinflammation 2019; 16:101. [PMID: 31092253 PMCID: PMC6521501 DOI: 10.1186/s12974-019-1497-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/30/2019] [Indexed: 01/13/2023] Open
Abstract
Background Blood-brain barrier (BBB) disruption and neuroinflammation are considered key mechanisms of pathogenic Escherichia coli invasion of the brain. However, the specific molecules involved in meningitic E. coli-induced BBB breakdown and neuroinflammatory response remain unclear. Our previous RNA-sequencing data from human brain microvascular endothelial cells (hBMECs) revealed two important host factors: platelet-derived growth factor-B (PDGF-B) and intercellular adhesion molecule-1 (ICAM-1), which were significantly upregulated in hBMECs after meningitic E. coli infection. Whether and how PDGF-B and ICAM-1 contribute to the development of E. coli meningitis are still unclear. Methods The western blot, real-time PCR, enzyme-linked immunosorbent assay, immunohistochemistry, and immunofluorescence were applied to verify the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in vivo and in vitro. Evan’s blue assay and electric cell-substrate impedance sensing assay were combined to identify the effects of PDGF-B on BBB permeability. The CRISPR/Cas9 technology, cell-cell adhesion assay, and electrochemiluminescence assay were used to investigate the role of ICAM-1 in neuroinflammation subversion. Results We verified the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in mouse as well as monolayer hBMECs models. Functionally, we showed that the increase of PDGF-B may directly enhance the BBB permeability by decreasing the expression of tight junction proteins, and the upregulation of ICAM-1 contributed to neutrophils or monocytes recruitment as well as neuroinflammation subversion in response to meningitic E. coli infection. Conclusions Our findings demonstrated the roles of PDGF-B and ICAM-1 in mediating bacterial-induced BBB damage as well as neuroinflammation, providing new concepts and potential targets for future prevention and treatment of bacterial meningitis.
Collapse
Affiliation(s)
- Rui-Cheng Yang
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xin-Yi Qu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Si-Yu Xiao
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Liang Li
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Bo-Jie Xu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Ji-Yang Fu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Yu-Jin Lv
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, Henan, China
| | - Nouman Amjad
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Chen Tan
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Kwang Sik Kim
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Huan-Chun Chen
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xiang-Ru Wang
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, Hubei, China. .,State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
27
|
Kurnick SA, Mannion AJ, Feng Y, Madden CM, Chamberlain P, Fox JG. Genotoxic Escherichia coli Strains Encoding Colibactin, Cytolethal Distending Toxin, and Cytotoxic Necrotizing Factor in Laboratory Rats. Comp Med 2019; 69:103-113. [PMID: 30902120 PMCID: PMC6464076 DOI: 10.30802/aalas-cm-18-000099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/13/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
Although many Escherichia coli strains are considered commensals in mammals, strains encoding the cyclomodulin genotoxins are associated with clinical and subclinical disease in the urogenital and gastrointestinal tracts, meningitis, and inflammatory disorders. These genotoxins include the polyketide synthase (pks) pathogenicity island, cytolethal distending toxin (cdt), and hemolysin-associated cytotoxic necrotizing factor (cnf). E. coli strains are not excluded from rodents housed under SPF conditions in academic or vendor facilities. This study isolated and characterized genotoxin-encoding E. coli from laboratory rats obtained from 4 academic institutions and 3 vendors. A total of 69 distinct E. coli isolates were cultured from feces, rectal swab, nares, or vaginal swab of 52 rats and characterized biochemically. PCR analysis for cyclomodulin genes and phylogroup was performed on all 69 isolates. Of the 69 isolates, 45 (65%) were positive for pks, 20/69 (29%) were positive for cdt, and 4 (6%) were positive for cnf. Colibactin was the sole genotoxin identified in 21 of 45 pks+ isolates (47%), whereas cdt or cnf was also present in the remaining 24 isolates (53%); cdt and cnf were never present together or without pks. All genotoxin-associated strains were members of pathogen-associated phylogroup B2. Fisher exact and χ² tests demonstrated significant differences in genotoxin prevalence and API code distribution with regard to vendor. Select E. coli isolates were characterized by HeLa cell in vitro cytotoxicity assays, serotyped, and whole-genome sequenced. All isolates encoding cyclomodulins induced megalocytosis. Serotypes corresponded with vendor origin and cyclomodulin composition, with the cnf+ serotype representing a known human uropathogen. Whole-genome sequencing confirmed the presence of complete pks, cdt, and hemolysin-cnf pathogenicity islands. These findings indicate that genotoxin-encoding E. coli colonize laboratory rats from multiple commercial vendors and academic institutions and suggest the potential to contribute to clinical disease and introduce confounding variables into experimental rat models.
Collapse
Affiliation(s)
- Susanna A Kurnick
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Anthony J Mannion
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Yan Feng
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Carolyn M Madden
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Paul Chamberlain
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts;,
| |
Collapse
|
28
|
Gao J, Li Y, Wan Y, Hu T, Liu L, Yang S, Gong Z, Zeng Q, Wei Y, Yang W, Zeng Z, He X, Huang SH, Cao H. A Novel Postbiotic From Lactobacillus rhamnosus GG With a Beneficial Effect on Intestinal Barrier Function. Front Microbiol 2019; 10:477. [PMID: 30923519 PMCID: PMC6426789 DOI: 10.3389/fmicb.2019.00477] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
It has long been known that probiotics can be used to maintain intestinal homeostasis and treat a number of gastrointestinal disorders, but the underlying mechanism has remained obscure. Recently, increasing evidence supports the notion that certain probiotic-derived components, such as bacteriocins, lipoteichoic acids, surface layer protein and secreted protein, have a similar protective role on intestinal barrier function as that of live probiotics. These bioactive components have been named 'postbiotics' in the most recent publications. We previously found that the Lactobacillus rhamnosus GG (LGG) culture supernatant is able to accelerate the maturation of neonatal intestinal defense and prevent neonatal rats from oral Escherichia coli K1 infection. However, the identity of the bioactive constituents has not yet been determined. In this study, using liquid chromatography-tandem mass spectrometry analysis, we identified a novel secreted protein (named HM0539 here) involved in the beneficial effect of LGG culture supernatant. HM0539 was recombinated, purified, and applied for exploring its potential bioactivity in vitro and in vivo. Our results showed that HM0539 exhibits a potent protective effect on the intestinal barrier, as reflected by enhancing intestinal mucin expression and preventing against lipopolysaccharide (LPS)- or tumor necrosis factor α (TNF-α)-induced intestinal barrier injury, including downregulation of intestinal mucin (MUC2), zonula occludens-1 (ZO-1) and disruption of the intestinal integrity. Using a neonatal rat model of E. coli K1 infection via the oral route, we verified that HM0539 is sufficient to promote development of neonatal intestinal defense and prevent against E. coli K1 pathogenesis. Moreover, we further extended the role of HM0539 and found it has potential to prevent dextran sulfate sodium (DSS)-induced colitis as well as LPS/D-galactosamine-induced bacterial translocation and liver injury. In conclusion, we identified a novel LGG postbiotic HM0539 which exerts a protective effect on intestinal barrier function. Our findings indicated that HM0539 has potential to become a useful agent for prevention and treatment of intestinal barrier dysfunction- related diseases.
Collapse
Affiliation(s)
- Jie Gao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yubin Li
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yu Wan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Tongtong Hu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Liting Liu
- Department of Medical Microbiology and Immunology, Dali University, Dali, China
| | - Shaojie Yang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zelong Gong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qing Zeng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yi Wei
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Weijun Yang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhijie Zeng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaolong He
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.,Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Qi-Deficiency Related Increases in Disease Susceptibility Are Potentially Mediated by the Intestinal Microbiota. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1304397. [PMID: 30425748 PMCID: PMC6218746 DOI: 10.1155/2018/1304397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/02/2018] [Indexed: 12/24/2022]
Abstract
Qi-deficiency (QX) is thought to promote the body's susceptibility to disease, but the underlying mechanism through which this occurs is not clear. We surveyed the traditional Chinese medicine constitution (TCMC) of healthy college students to identify those that were PH (balanced TCMC constitution) and QX (unbalanced TCMC constitution). We then used high-throughput sequencing of the 16SrRNA V3-4 region in fecal microbiota samples to identify differences between those obtained from PH and QX individuals. Our results demonstrated that the alpha diversity of QX samples was significantly lower than that of PH samples (p < 0.05) and that beta diversity was remarkably different in QX and PH samples. Four and 122 bacterial taxa were significantly overrepresented in QX and PH groups, respectively. The genera Sphingobium, Clostridium, and Comamonas were enriched in the QX group and had a certain pathogenic role. The QX group also showed a statistically significant lack of probiotics and anti-inflammatory bacteria such as Bifidobacterium and Bdellovibrio. The functional potential of QX bacterial taxa was reduced in fatty acid metabolism and butanoate metabolism. We contend that the imbalanced intestinal microbiota in QX and the following functional changes in metabolism influence immunity and energy metabolism, which could increase susceptibility to disease.
Collapse
|
30
|
Xu C, Guo Y, Qiao L, Ma L, Cheng Y, Roman A. Biogenic Synthesis of Novel Functionalized Selenium Nanoparticles by Lactobacillus casei ATCC 393 and Its Protective Effects on Intestinal Barrier Dysfunction Caused by Enterotoxigenic Escherichia coli K88. Front Microbiol 2018; 9:1129. [PMID: 29967593 PMCID: PMC6015882 DOI: 10.3389/fmicb.2018.01129] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/14/2018] [Indexed: 11/13/2022] Open
Abstract
Selenium (Se) is an essential element for human and animal health. Biogenic selenium nanoparticles (SeNPs) by microorganism possess unique physical and chemical properties and biological activities compared with inorganic Se and organic Se. The study was conducted to investigate the mainly biological activities of SeNPs by Lactobacillus casei ATCC 393 (L. casei 393). The results showed that L. casei 393 transformed sodium selenite to red SeNPs with the size of 50–80 nm, and accumulated them intracellularly. L. casei 393-SeNPs promoted the growth and proliferation of porcine intestinal epithelial cells (IPEC-J2), human colonic epithelial cells (NCM460), and human acute monocytic leukemia cell (THP-1)-derived macrophagocyte. L. casei 393-SeNPs significantly inhibited the growth of human liver tumor cell line-HepG2, and alleviated diquat-induced IPEC-J2 oxidative damage. Moreover, in vivo and in vitro experimental results showed that administration with L. casei 393-SeNPs protected against Enterotoxigenic Escherichia coli K88 (ETEC K88)-caused intestinal barrier dysfunction. ETEC K88 infection-associated oxidative stress (glutathione peroxidase activity, total superoxide dismutase activity, total antioxidant capacity, and malondialdehyde) was ameliorated in L. casei 393-SeNPs-treated mice. These findings suggest that L. casei 393-SeNPs with no cytotoxicity play a key role in maintaining intestinal epithelial integrity and intestinal microflora balance in response to oxidative stress and infection.
Collapse
Affiliation(s)
- Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yu Guo
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Li Ma
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yiyi Cheng
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Alexandra Roman
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|