1
|
Sulaimi F, Ong TSK, Tang ASP, Quek J, Pillay RM, Low DT, Lee CKL, Siah KTH, Ng QX. Risk factors for developing irritable bowel syndrome: systematic umbrella review of reviews. BMC Med 2025; 23:103. [PMID: 39985070 PMCID: PMC11846330 DOI: 10.1186/s12916-025-03930-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a debilitating disorder affecting 4-9% of the global population. It is a multifaceted disorder with complex and varied causes. This review aims to consolidate the evidence regarding IBS risk factors by examining existing systematic reviews and meta-analyses, covering potential genetic, immunological, psychological, and dietary causes. METHODS Systematic literature searches were conducted in MEDLINE, Embase and Cochrane library databases. Study selection and data extraction were conducted independently by four authors, with discrepancies resolved by consensus with a senior author. Systematic reviews examining risk factors of IBS development were eligible for review. Results were narratively synthesized. Quality of reviews were analysed using AMSTAR 2, and evidence were appraised using GRADE methodology. RESULTS A total of 69 systematic reviews were included in this study. Most reviews were of "critically low" quality, while the remaining were "low" quality. Common shortcomings included the absence of a list of excluded studies with justifications for their exclusion and inadequate consideration of the risk of bias in individual studies. Eight major categories of risk factors for IBS identified were as follows: dietary, genetic, environmental, psychological, gut microbiome, socio-economic, physiological, and pathological, albeit overlaps exist. The most frequently reported risk factors for IBS development were female gender and anxiety disorders, with overall GRADE evaluation of "low"; depression and gastroenteritis, with overall GRADE evaluation of "moderate". CONCLUSIONS Clinical practice should prioritize recognition of these risk factors. Future reviews should improve their reporting of results based on the PRISMA guidelines, to enhance the quality of research in this field. PROTOCOL REGISTRATION PROSPERO CRD42023493739.
Collapse
Affiliation(s)
- Farisah Sulaimi
- School of Medical Sciences, Wallace Wurth Building, University of New South Wales, Sydney, NSW, Australia
| | - Timothy Sheng Khai Ong
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ansel Shao Pin Tang
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jingxuan Quek
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Renish M Pillay
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Damien Tianle Low
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charisse Kai Ling Lee
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kewin Tien Ho Siah
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology & Hepatology, University Medicine Cluster, National University Hospital, Singapore, Singapore
| | - Qin Xiang Ng
- NUS Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore.
- SingHealth Duke-NUS Global Health Institute, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
2
|
Fneish FH, Abd El Galil KH, Domiati SA. Evaluation of Single and Multi-Strain Probiotics with Gentamicin Against E. coli O157:H7: Insights from In Vitro and In Vivo Studies. Microorganisms 2025; 13:460. [PMID: 40005825 PMCID: PMC11858083 DOI: 10.3390/microorganisms13020460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
The emergence of antibiotic-resistant food-borne pathogens, especially Escherichia coli O157:H7, highlights the urgent need for innovative treatment strategies, particularly in light of rising resistances and the ongoing controversy surrounding antibiotic use in response to E. coli O157:H7 infections. To address this issue, we explored the potential of single- and multi-strain probiotics, both independently and in combination with gentamicin, through a series of in vitro and in vivo experiments. In vitro, gentamicin alone produced a mean inhibition zone of 12.9 ± 2.27 mm against E. coli O157:H7. The combination of gentamicin with single-strain probiotics (P1) increased the inhibition zone to 16.5 ± 2.24 mm (p < 0.05), while the combination with multi-strain probiotics (P2) resulted in the largest inhibition zone of 19 ± 2.8 mm (p < 0.05). In vivo, mice infected with E. coli O157:H7 and treated with P2, gentamicin (G), or their combination (G+P2), achieved 100% survival, no pathological symptoms, and full weight recovery within seven days. Conversely, mice treated with P1 or G+P1 exhibited lower survival rates (71.4% and 85%, respectively) and slower weight recovery. Hematological parameters improved across all groups, but kidney function analysis showed significantly higher serum creatinine levels in the P1, G, G+P1, and G+P2 groups compared to the P2 group (P1: 0.63 ± 0.15 mg/dL; G: 0.34 ± 0.09 mg/dL; G+P1: 0.53 ± 0.19 mg/dL; G+P2: 0.5 ± 0.23 mg/dL vs. P2: 0.24 ± 0.2 mg/dL). Histological analysis showed better intestinal and kidney tissue recovery in the P2 group, while the P1 and G+P1 groups exhibited abnormal ileal structures and severe cortical bleeding. These findings highlight the promise of multi-strain probiotics, alone or in conjunction with antibiotics, as a therapeutic strategy for E. coli O157:H7 infections. However, the nephrotoxicity associated with gentamicin co-administration remains a limitation, warranting further studies to optimize this approach.
Collapse
Affiliation(s)
- Fatima H. Fneish
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Beirut Arab University, Riad El Solh P.O. Box 11-5020, Beirut 11072809, Lebanon
| | - Khaled H. Abd El Galil
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt;
| | - Souraya A. Domiati
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, Beirut 11072809, Lebanon;
| |
Collapse
|
3
|
Bowser S, Chapartegui-González I, Torres AG. Fecal microbiome alterations of mice following immunization with gold nanoparticle vaccines against enterohemorrhagic Escherichia coli. Gut Pathog 2024; 16:75. [PMID: 39702387 DOI: 10.1186/s13099-024-00670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Enterohemorrhagic Escherichia coli (EHEC), a group of enteric pathogenic bacteria that is a major cause of human diarrheal disease, must interact with the diverse intestinal microbiome during colonization and subsequently overcome the environmental challenges to survive and cause disease. While this relationship, and how the microbiome modulates infection of EHEC, has been studied, it is less understood how the microbiome is impacted during treatment for an EHEC infection. One area that is notably lacking in knowledge is how vaccination can impact the intestinal microbiome composition, and therefore, influence vaccine efficacy. We previously developed vaccine formulations consisting of gold nanoparticles (AuNPs) conjugated to various EHEC antigens and tested them in mice models using both EHEC and its murine counterpart Citrobacter rodentium. The goal of this study was to evaluate the relationship between these EHEC vaccines and their effects on the gut microbiome. RESULTS We found that immunization with the vaccines or adjuvant-only control did not lead to major alterations in the composition of the fecal microbiome; however, there were measurable variations in individual mice within the same vaccine group housed in separate cages. Also, immunization with one vaccine (AuNP-EscC) prevented both a decrease in the diversity of the fecal microbiome and an increase in detectable C. rodentium following infection compared to control animals. CONCLUSIONS Overall, our small study argues in favor of evaluating the intestinal microbiome during vaccine development not just for EHEC, but for other enteric pathogens.
Collapse
Affiliation(s)
- Sarah Bowser
- Department of Microbiology and Immunology, Galveston, TX, USA
| | - Itziar Chapartegui-González
- Department of Microbiology and Immunology, Galveston, TX, USA
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 52, Sweden
| | - Alfredo G Torres
- Department of Microbiology and Immunology, Galveston, TX, USA.
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
4
|
Bowser S, Chapartegui-González I, Torres AG. Fecal Microbiome Alterations of Mice Following Immunization with Gold Nanoparticle Vaccines Against Enterohemorrhagic Escherichia coli. RESEARCH SQUARE 2024:rs.3.rs-5146579. [PMID: 39649169 PMCID: PMC11623765 DOI: 10.21203/rs.3.rs-5146579/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Background Enterohemorrhagic Escherichia coli (EHEC), a group of enteric pathogenic bacteria that is a major cause of human diarrheal disease, must interact with the diverse intestinal microbiome during colonization and subsequently overcome the environmental challenges to survive and cause disease. While this relationship, and how the microbiome modulates infection of EHEC, has been studied, it is less understood how the microbiome is impacted during treatment for an EHEC infection. One area that is notably lacking in knowledge is how vaccination can impact the intestinal microbiome composition, and therefore, influence vaccine efficacy. We previously developed vaccine formulations consisting of gold nanoparticles (AuNPs) conjugated to various EHEC antigens and tested them in small animal infection models using both EHEC and its murine counterpart Citrobacter rodentium. The goal of this study was to evaluate the relationship between these EHEC vaccines and their effects on the gut microbiome. Results We found that immunization with the vaccines or adjuvant-only control did not lead to major alterations in the composition of the fecal microbiome; however, there were measurable variations in individual mice within the same vaccine group housed in separate cages. Finally, immunization with one vaccine (AuNP-EscC) did prevent a decrease in the diversity of the fecal microbiome and an increase in detectable C. rodentium following infection compared to the control animals. Conclusions Overall, our small study argues in favor of evaluating the intestinal microbiome during vaccine development not just for EHEC, but for other enteric pathogens as well.
Collapse
|
5
|
Nagao I, Kawasaki M, Goyama T, Kim HJ, Call DR, Ambrosini YM. Enterohemorrhagic Escherichia coli (EHEC) disrupts intestinal barrier integrity in translational canine stem cell-derived monolayers. Microbiol Spectr 2024; 12:e0096124. [PMID: 39162490 PMCID: PMC11448187 DOI: 10.1128/spectrum.00961-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/30/2024] [Indexed: 08/21/2024] Open
Abstract
This study addresses the gap in translatable in vitro models for investigating Enterohemorrhagic E. coli (EHEC) infections, particularly relevant to both canine and human health. EHEC is known to induce acute colitis in dogs, leading to symptoms like hemorrhagic diarrhea and hemolytic uremic syndrome, similar to those observed in humans. However, understanding the pathophysiology and developing treatment strategies have been challenging due to the lack of effective models that replicate the clinical disease caused by EHEC in both species. Our approach involved the development of colonoid-derived monolayers using intestinal tissues from healthy, client-owned dogs. These monolayers were exposed to EHEC, and the impact of EHEC was assessed through several techniques, including trans-epithelial electrical resistance (TEER) measurement, immunofluorescence staining for junction proteins and mucus, and scanning electron microscopy for morphological analysis. Modified culture with saline, which was intended to prevent bacterial overgrowth, maintained barrier integrity and cell differentiation. EHEC infection led to significant decreases in TEER and ZO-1 expression, but not in E-cadherin levels or mucus production. In addition, EHEC elicited a notable increase in tumor necrosis factor-alpha production, highlighting its distinct impact on canine intestinal epithelial cells compared to non-pathogenic E. coli. These findings closely replicate in vivo observations in dogs and humans with EHEC enteropathy, validating the canine colonoid-derived monolayer system as a translational model to study host-pathogen interactions in EHEC and potentially other clinically significant enteric pathogens. IMPORTANCE This study develops a new model to better understand Enterohemorrhagic E. coli (EHEC) infections, a serious bacterial disease affecting both dogs and humans, characterized by symptoms such as hemorrhagic diarrhea and hemolytic uremic syndrome. Traditional research models have fallen short of mimicking how this disease manifests in patients. Our research used intestinal tissues from healthy dogs to create layers of cells, known as colonoid-derived monolayers, which we then exposed to EHEC. We assessed the damage caused by the bacteria using several techniques, observing significant changes similar to those seen in actual cases of the disease. The model proved effective in replicating the interaction between the host and the pathogen, marking an important step toward understanding EHEC's effects and developing treatments. This canine colonoid-derived monolayer system not only bridges a crucial gap in current research but also offers a promising platform for studying other enteric pathogens affecting both canine and human health.
Collapse
Affiliation(s)
- Itsuma Nagao
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Minae Kawasaki
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Takashi Goyama
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Hyun Jung Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Douglas R. Call
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Yoko M. Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
6
|
Yu T, Chen D, Qi H, Lin L, Tang Y. Resolvins protect against diabetes-induced colonic oxidative stress, barrier dysfunction, and associated diarrhea via the HO-1 pathway. Biofactors 2024; 50:967-979. [PMID: 38485285 DOI: 10.1002/biof.2049] [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: 07/03/2023] [Accepted: 12/26/2023] [Indexed: 10/04/2024]
Abstract
Diabetes is associated with increased oxidative stress, leading to altered tight junction formation and increased apoptosis in colonic epithelial cells. These changes may lead to intestinal barrier dysfunction and corresponding gastrointestinal symptoms in patients with diabetes, including diarrhea. The aim of this study was to characterize the effect and mechanism of Resolvin D1 (RvD1) on diabetes-induced oxidative stress and barrier disruption in the colon. Mice with streptozotocin-induced diabetes were treated with RvD1 for 2 weeks, then evaluated for stool frequency, stool water content, gut permeability, and colonic transepithelial electrical resistance as well as production of reactive oxygen species (ROS), apoptosis, and expression of tight junction proteins Zonula Occludens 1 (ZO-1) and occludin. The same parameters were assessed in human colonoid cultures subjected to elevated glucose. We found that RvD1 treatment did not affect blood glucose, but normalized stool water content and prevented intestinal barrier dysfunction, epithelial oxidative stress, and apoptosis. RvD1 also restored ZO-1 and occludin expression in diabetic mice. RvD1 treatment increased phosphorylation of Akt and was accompanied by a 3.5-fold increase in heme oxygenase-1 (HO-1) expression in the epithelial cells. The protective effects of RvD1 were blocked by ZnPP, a competitive inhibitor of HO-1. Similar findings were observed in RvD1-treated human colonoid cultures subjected to elevated glucose. In conclusion, Oxidative stress in diabetes results in mucosal barrier dysfunction, contributing to the development of diabetic diarrhea. Resolvins prevent ROS-mediated mucosal injury and protect gut barrier function by intracellular PI3K/Akt activation and subsequent HO-1 upregulation in intestinal epithelial cells. These actions result in normalizing stool frequency and stool water content in diabetic mice, suggesting that resolvins may be useful in the treatment of diabetic diarrhea.
Collapse
Affiliation(s)
- Ting Yu
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province, China
| | - Die Chen
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province, China
| | - Hongyan Qi
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province, China
| | - Lin Lin
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province, China
| | - Yurong Tang
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province, China
| |
Collapse
|
7
|
Cheng R, Wang J, Wu Q, Peng P, Liao G, Luo X, Liang Z, Huang J, Qin M. The Predictive Value of Serum DAO, HDC, and MMP8 for the Gastrointestinal Injury in the Early Stage of Acute Pancreatitis in an Animal Model and a Clinical Study. Int J Gen Med 2024; 17:1937-1948. [PMID: 38736673 PMCID: PMC11088402 DOI: 10.2147/ijgm.s461352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose This study was aimed at exploring the use of the acute gastrointestinal injury (AGI) grade and sensitive biomarkers to investigate gastrointestinal (GI) injury in early stage of acute pancreatitis (AP). Patients and Methods The AGI grade was used to evaluate intestinal function. Any GI injury above grade I (grades II-IV) was considered as severe. An AP rat model was created by retrograde injection of 4% sodium taurocholate. The pancreatic and intestinal histopathology scores were calculated by hematoxylin-eosin staining. Human and rat sera were assessed using ELISA. Tight junction (TJ) proteins were detected by Western blotting. Results In clinical study, the GI injury rate in mild acute pancreatitis (MAP), moderate severe acute pancreatitis (MSAP), and severe acute pancreatitis (SAP) groups was 26.8%, 78.4%, and 94.8%, respectively (P < 0.05). Diamine oxidase (DAO), histidine decarboxylase (HDC), and matrix metalloproteinase 8 (MMP8) serum levels were higher in AP patients than in healthy people (P < 0.05). Patients with GI injury had higher serum levels of DAO, HDC, and MMP8 than those without GI injury (P < 0.05). In animal experiments, the serum levels of DAO, HDC, and MMP8 were higher in the AP group than in normal and sham-operated (SO) groups (P < 0.05). The expressions of tricellulin, claudin-1, ZO-1, and occludin were significantly lower in the AP group than in normal and SO groups (P < 0.05). Conclusion The serum levels of DAO, HDC, and MMP8 are novel biomarkers of GI injury in the early stage of AP; their elevation indicates the development of GI injury in AP. The intestinal TJ disruption may be a primary mechanism of GI injury and requires more in-depth research.
Collapse
Affiliation(s)
- Ruoxi Cheng
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Jie Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Qing Wu
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Peng Peng
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Guolin Liao
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Xiuping Luo
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Zhihai Liang
- Department of Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Jiean Huang
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Mengbin Qin
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| |
Collapse
|
8
|
Kawasaki M, Ambrosini YM. Differential Colonization and Mucus Ultrastructure Visualization in Bovine Ileal and Rectal Organoid-Derived Monolayers Exposed to Enterohemorrhagic Escherichia coli. Int J Mol Sci 2024; 25:4914. [PMID: 38732126 PMCID: PMC11084217 DOI: 10.3390/ijms25094914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.
Collapse
Affiliation(s)
| | - Yoko M. Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
9
|
Vasilopoulos S, Giannenas I, Mellidou I, Stylianaki I, Antonopoulou E, Tzora A, Skoufos I, Athanassiou CG, Papadopoulos E, Fortomaris P. Diet replacement with whole insect larvae affects intestinal morphology and microbiota of broiler chickens. Sci Rep 2024; 14:6836. [PMID: 38514719 PMCID: PMC10957974 DOI: 10.1038/s41598-024-54184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
Insect-based diets are gaining interest as potential ingredients in improving poultry gut health. This study assessed the dietary treatment with whole dried Tenebrio molitor larvae (TM) on broiler chickens' gut microbiota and morphology. 120 Ross-308 broilers received treated diets with 5% (TM5) and 10% (TM10) replacement ratio in a 35-day trial. Intestinal histomorphometry was assessed, as well as claudin-3 expression pattern and ileal and caecal digesta for microbial community diversity. Null hypothesis was tested with two-way ANOVA considering the intestinal segment and diet as main factors. The TM5 group presented higher villi in the duodenum and ileum compared to the other two (P < 0.001), while treated groups showed shallower crypts in the duodenum (P < 0.001) and deeper in the jejunum and ileum than the control (P < 0.001). Treatments increased the caecal Firmicutes/Bacteroidetes ratio and led to significant changes at the genus level. While Lactobacilli survived in the caecum, a significant reduction was evident in the ileum of both groups, mainly owed to L. aviarius. Staphylococci and Methanobrevibacter significantly increased in the ileum of the TM5 group. Results suggest that dietary supplementation with whole dried TM larvae has no adverse effect on the intestinal epithelium formation and positively affects bacterial population richness and diversity.
Collapse
Affiliation(s)
- Stylianos Vasilopoulos
- Laboratory of Nutrition, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece.
| | - Ilias Giannenas
- Laboratory of Nutrition, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece
| | - Ifigeneia Mellidou
- Institute of Plant Breeding and Genetic Resources, Hellenic Agricultural Organization-DIMITRA, 57001, Thessaloníki, Greece
| | - Ioanna Stylianaki
- Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece
| | - Efthimia Antonopoulou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100, Arta, Greece
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, 47100, Arta, Greece
| | - Christos G Athanassiou
- Laboratory of Entomology and Agricultural Zoology, Department of Agriculture, Crop Production and Rural Environment, University of Thessaly, Phytokou Str., 38446, N. Ionia, Volos, Greece
| | - Elias Papadopoulos
- Laboratory of Parasitology and Parasitic Diseases, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece
| | - Paschalis Fortomaris
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 54124, Thessaloníki, Greece
| |
Collapse
|
10
|
Zhuo W, Zhao Y, Zhao X, Yao Z, Qiu X, Huang Y, Li H, Shen J, Zhu Z, Li T, Li S, Huang Q, Zhou R. Enteropathogenic Escherichia coli is a predominant pathotype in healthy pigs in Hubei Province of China. J Appl Microbiol 2023; 134:lxad260. [PMID: 37962953 DOI: 10.1093/jambio/lxad260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/29/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023]
Abstract
AIM This study aims to investigate the prevalence of intestinal pathogenic Escherichia coli (InPEC) in healthy pig-related samples and evaluate the potential virulence of the InPEC strains. METHODS AND RESULTS A multiplex PCR method was established to identify different pathotypes of InPEC. A total of 800 rectal swab samples and 296 pork samples were collected from pig farms and slaughterhouses in Hubei province, China. From these samples, a total of 21 InPEC strains were isolated, including 19 enteropathogenic E. coli (EPEC) and 2 shiga toxin-producing E. coli (STEC) strains. By whole-genome sequencing and in silico typing, it was shown that the sequence types and serotypes were diverse among the strains. Antimicrobial susceptibility assays showed that 90.48% of the strains were multi-drug resistant. The virulence of the strains was first evaluated using the Galleria mellonella larvae model, which showed that most of the strains possessed medium to high pathogenicity. A moderately virulent EPEC isolate was further selected to characterize its pathogenicity using a mouse model, which suggested that it could cause significant diarrhea. Bioluminescence imaging (BLI) was then used to investigate the colonization dynamics of this EPEC isolate, which showed that the EPEC strain could colonize the mouse cecum for up to 5 days.
Collapse
Affiliation(s)
- Wenxiao Zhuo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Zhao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xianglin Zhao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhiming Yao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuxiu Qiu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yaxue Huang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huaixia Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Shen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhihao Zhu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Tingting Li
- Hubei Animal Disease Prevention and Control Center, Wuhan 430070, China
| | - Shaowen Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qi Huang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, College of Veterinary Medicine, Wuhan 430070, China
- International Research Center for Animal Disease (Ministry of Science & Technology of China), College of Veterinary Medicine, Wuhan 430070, China
| | - Rui Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, College of Veterinary Medicine, Wuhan 430070, China
- International Research Center for Animal Disease (Ministry of Science & Technology of China), College of Veterinary Medicine, Wuhan 430070, China
- The HZAU-HVSEN Research Institute, Wuhan 430042, China
| |
Collapse
|
11
|
Tóth Š, Fagová Z, Holodová M, Zeidan D, Hartel P, Čurgali K, Mechírová E, Maretta M, Nemcová R, Gancarčíková S, Danková M. Influence of Escherichia coli infection on intestinal mucosal barrier integrity of germ-free piglets. Life Sci 2023; 331:122036. [PMID: 37633417 DOI: 10.1016/j.lfs.2023.122036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
AIMS We focused on investigating the influence of Escherichia coli (E. coli) on the intestinal barrier. MATERIAL AND METHODS We studied changes in the distribution and secretory activities of goblet cells and enteroendocrine cells (EECs), as well as changes in the population of mast cells (MCs) in the jejunal and colonic mucosa of germ-free (GF) piglets as a healthy control group and GF piglets whose intestines were colonised with E. coli bacteria on day 5. KEY FINDINGS The results suggest that the colon of GF piglets is more resistant and less prone to coliform bacterial infection compared to the jejunum. This can be confirmed by a lower degree of histopathological injury index as well as an improvement of the morphometric parameters of the colonic mucosa, together with a significantly increased (p < 0.05) expression of MUC1/EMA, and ZO-3. We also observed a significant decrease in the population of activated MCs (p < 0.001) and EECs (p < 0.001). These findings may indicate a rapid response and better preparation of the intestinal barrier for possible pathological attacks and the subsequent development of mucosal lesions during the development and progression of the intestinal diseases. SIGNIFICANCE To date, gut-targeted therapeutic approaches that can modulate bacterial translocation and chronic inflammation are still in their infancy but represent one of the most promising areas of research for the development of new effective treatments or clinical strategies in the future. Therefore, a better understanding of these processes can significantly contribute to the development of these targeted strategies for disease prevention and treatment.
Collapse
Affiliation(s)
- Štefan Tóth
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Zuzana Fagová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Monika Holodová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Dema Zeidan
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Patrick Hartel
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Kristína Čurgali
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Eva Mechírová
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Histology and Embryology, Šrobárova 2, 040 01 Košice, Slovak Republic
| | - Milan Maretta
- Pavol Jozef Šafárik University, Faculty of Medicine, Department of Neurology and L. Pasteur University Hospital, Trieda SNP 1, 040 01 Košice, Slovak Republic
| | - Radomíra Nemcová
- University of Veterinary Medicine and Pharmacy in Košice, Department of Microbiology and Immunology, Komenského 73, 041 70 Košice, Slovak Republic
| | - Soňa Gancarčíková
- University of Veterinary Medicine and Pharmacy in Košice, Department of Microbiology and Immunology, Komenského 73, 041 70 Košice, Slovak Republic
| | - Marianna Danková
- Comenius University in Bratislava, Faculty of Medicine, Institute of Histology and Embryology, Sasinkova 4, 811 04 Bratislava, Slovak Republic.
| |
Collapse
|
12
|
Lupu VV, Ghiciuc CM, Stefanescu G, Mihai CM, Popp A, Sasaran MO, Bozomitu L, Starcea IM, Adam Raileanu A, Lupu A. Emerging role of the gut microbiome in post-infectious irritable bowel syndrome: A literature review. World J Gastroenterol 2023; 29:3241-3256. [PMID: 37377581 PMCID: PMC10292139 DOI: 10.3748/wjg.v29.i21.3241] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/04/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Post-infectious irritable bowel syndrome (PI-IBS) is a particular type of IBS, with symptom onset after an acute episode of infectious gastroenteritis. Despite infectious disease resolution and clearance of the inciting pathogen agent, 10% of patients will develop PI-IBS. In susceptible individuals, the exposure to pathogenic organisms leads to a marked shift in the gut microbiota with prolonged changes in host-microbiota interactions. These changes can affect the gut-brain axis and the visceral sensitivity, disrupting the intestinal barrier, altering neuromuscular function, triggering persistent low inflammation, and sustaining the onset of IBS symptoms. There is no specific treatment strategy for PI-IBS. Different drug classes can be used to treat PI-IBS similar to patients with IBS in general, guided by their clinical symptoms. This review summarizes the current evidence for microbial dysbiosis in PI-IBS and analyzes the available data regarding the role of the microbiome in mediating the central and peripheral dysfunctions that lead to IBS symptoms. It also discusses the current state of evidence on therapies targeting the microbiome in the management of PI-IBS. The results of microbial modulation strategies used in relieving IBS symptomatology are encouraging. Several studies on PI-IBS animal models reported promising results. However, published data that describe the efficacy and safety of microbial targeted therapy in PI-IBS patients are scarce. Future research is required.
Collapse
Affiliation(s)
- Vasile Valeriu Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Cristina Mihaela Ghiciuc
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Gabriela Stefanescu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | | | - Alina Popp
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest 020021, Romania
| | - Maria Oana Sasaran
- Faculty of General Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures 540142, Romania
| | - Laura Bozomitu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Iuliana Magdalena Starcea
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Anca Adam Raileanu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Ancuta Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| |
Collapse
|
13
|
Krsek D, Yara DA, Hrbáčková H, Daniel O, Mančíková A, Schüller S, Bielaszewska M. Translocation of outer membrane vesicles from enterohemorrhagic Escherichia coli O157 across the intestinal epithelial barrier. Front Microbiol 2023; 14:1198945. [PMID: 37303786 PMCID: PMC10248468 DOI: 10.3389/fmicb.2023.1198945] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Outer membrane vesicles (OMVs) carrying virulence factors of enterohemorrhagic Escherichia coli (EHEC) are assumed to play a role in the pathogenesis of life-threatening hemolytic uremic syndrome (HUS). However, it is unknown if and how OMVs, which are produced in the intestinal lumen, cross the intestinal epithelial barrier (IEB) to reach the renal glomerular endothelium, the major target in HUS. We investigated the ability of EHEC O157 OMVs to translocate across the IEB using a model of polarized Caco-2 cells grown on Transwell inserts and characterized important aspects of this process. Using unlabeled or fluorescently labeled OMVs, tests of the intestinal barrier integrity, inhibitors of endocytosis, cell viability assay, and microscopic techniques, we demonstrated that EHEC O157 OMVs translocated across the IEB. OMV translocation involved both paracellular and transcellular pathways and was significantly increased under simulated inflammatory conditions. In addition, translocation was not dependent on OMV-associated virulence factors and did not affect viability of intestinal epithelial cells. Importantly, translocation of EHEC O157 OMVs was confirmed in human colonoids thereby supporting physiological relevance of OMVs in the pathogenesis of HUS.
Collapse
Affiliation(s)
- Daniel Krsek
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | | | - Hana Hrbáčková
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Ondřej Daniel
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Andrea Mančíková
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Martina Bielaszewska
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| |
Collapse
|
14
|
Sun S, Xu Z, Hu H, Zheng M, Zhang L, Xie W, Sun L, Liu P, Li T, Zhang L, Chen M, Zhu X, Liu M, Yang Y, Zhou J. The Bacillus cereus toxin alveolysin disrupts the intestinal epithelial barrier by inducing microtubule disorganization through CFAP100. Sci Signal 2023; 16:eade8111. [PMID: 37192300 DOI: 10.1126/scisignal.ade8111] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 04/18/2023] [Indexed: 05/18/2023]
Abstract
Bacillus cereus is a Gram-positive bacterium that mainly causes self-limiting emetic or diarrheal illness but can also cause skin infections and bacteremia. Symptoms of B. cereus ingestion depend on the production of various toxins that target the gastric and intestinal epithelia. From a screen of bacterial isolates from human stool samples that compromised intestinal barrier function in mice, we identified a strain of B. cereus that disrupted tight and adherens junctions in the intestinal epithelium. This activity was mediated by the pore-forming exotoxin alveolysin, which increased the production of the membrane-anchored protein CD59 and of cilia- and flagella-associated protein 100 (CFAP100) in intestinal epithelial cells. In vitro, CFAP100 interacted with microtubules and promoted microtubule polymerization. CFAP100 overexpression stabilized microtubules in intestinal epithelial cells, leading to disorganization of the microtubule network and perturbation of tight and adherens junctions. The disruption of cell junctions by alveolysin depended on the increase in CFAP100, which in turn depended on CD59 and the activation of PI3K-AKT signaling. These findings demonstrate that, in addition to forming membrane pores, B. cereus alveolysin can permeabilize the intestinal epithelium by disrupting epithelial cell junctions in a manner that is consistent with intestinal symptoms and may allow the bacteria to escape the intestine and cause systemic infections. Our results suggest the potential value of targeting alveolysin or CFAP100 to prevent B. cereus-associated intestinal diseases and systemic infections.
Collapse
Affiliation(s)
- Shuang Sun
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Zhaoyang Xu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Haijie Hu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Manxi Zheng
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liang Zhang
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Wei Xie
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Lei Sun
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Peiwei Liu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Tianliang Li
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liangran Zhang
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Min Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Liu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jun Zhou
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
15
|
Huang FC, Huang SC. The Pivotal Role of Aryl Hydrocarbon Receptor-Regulated Tight Junction Proteins and Innate Immunity on the Synergistic Effects of Postbiotic Butyrate and Active Vitamin D3 to Defense against Microbial Invasion in Salmonella Colitis. Nutrients 2023; 15:305. [PMID: 36678175 PMCID: PMC9860786 DOI: 10.3390/nu15020305] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Our recent report illustrated the unitedly advantageous effects of postbiotic butyrate on active vitamin D3 (VD3)-orchestrated innate immunity in Salmonella colitis. There is growing awareness that aryl hydrocarbon receptor (AhR) can regulate intestinal immunity and barrier function, through modulating cecal inflammation and junction proteins expression. Hence, we researched the participation of AhR-regulated tight junction functions on the united effects of butyrate and VD3 on intestinal defense to Salmonella infection. Salmonella colitis model were elicited by oral gavage with 1 × 108 CFU of a S. typhimurium wild-type strain SL1344 in C57BL/6 mice. Before and after the colitis generation, mice were fed with butyrate and/or VD3 by oral gavage in the absence or presence of intraperitoneal injection of AhR inhibitor for 4 and 7 days, respectively. We observed that butyrate and VD3 could concert together to reduce the invasion of Salmonella in colitis mice by enhancing cecal cytokines and antimicrobial peptides expression and reducing zonulin and claudin-2 protein expressions in mucosal stain, compared to single treatment, which were counteracted by AhR inhibitor. It implies that AhR is involved in the united effects of butyrate and VD3 on the intestinal defense to Salmonella infection in colitis mice. This study discloses the promising alternative therapy of combining postbiotic and VD3 for invasive Salmonellosis and the pivotal role of AhR pathway.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shun-Chen Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
16
|
Ren Z, Fan H, Deng H, Yao S, Jia G, Zuo Z, Hu Y, Shen L, Ma X, Zhong Z, Deng Y, Yao R, Deng J. Effects of dietary protein level on small intestinal morphology, occludin protein, and bacterial diversity in weaned piglets. Food Sci Nutr 2022; 10:2168-2201. [PMID: 35844902 PMCID: PMC9281955 DOI: 10.1002/fsn3.2828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 11/30/2022] Open
Abstract
Due to the physiological characteristics of piglets, the morphological structure and function of the small intestinal mucosa change after weaning, which easily leads to diarrhea in piglets. The aim of this study was to investigate effects of crude protein (CP) levels on small intestinal morphology, occludin protein expression, and intestinal bacteria diversity in weaned piglets. Ninety-six weaned piglets (25 days of age) were randomly divided into four groups and fed diets containing 18%, 20%, 22%, and 24% protein. At 6, 24, 48, 72, and 96 h, changes in mucosal morphological structure, occludin mRNA, and protein expression and in the localization of occludin in jejunal and ileal tissues were evaluated. At 6, 24, and 72 h, changes in bacterial diversity and number of the ileal and colonic contents were analyzed. Results showed that structures of the jejunum and the ileum of piglets in the 20% CP group were intact. The expression of occludin mRNA and protein in the small intestine of piglets in the 20% CP group were significantly higher than those in the other groups. As the CP level increased, the number of pathogens, such as Clostridium difficile and Escherichia coli, in the intestine increased, while the number of beneficial bacteria, such as Lactobacillus, Bifidobacterium, and Roseburia, decreased. It is concluded that maintaining the CP level at 20% is beneficial to maintaining the small intestinal mucosal barrier and its absorption function, reducing the occurrence of diarrhea, and facilitating the growth and development of piglets.
Collapse
Affiliation(s)
- Zhihua Ren
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Haoyue Fan
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Huidan Deng
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Shuhua Yao
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Guilin Jia
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Zhicai Zuo
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Yanchun Hu
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Liuhong Shen
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Xiaoping Ma
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Zhijun Zhong
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Youtian Deng
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Renjie Yao
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| | - Junliang Deng
- College of Veterinary MedicineSichuan Agricultural UniversityYa’anChina
- Sichuan Province Key Laboratory of Animal Disease & Human HealthYa’anChina
- Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceYa’anChina
| |
Collapse
|
17
|
Ismail HTH. The ameliorative efficacy of Thymus vulgaris essential oil against Escherichia coli O157:H7-induced hematological alterations, hepatorenal dysfunction and immune-inflammatory disturbances in experimentally infected rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:41476-41491. [PMID: 35088282 DOI: 10.1007/s11356-022-18896-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/23/2022] [Indexed: 06/14/2023]
Abstract
The purpose of the present study was to evaluate the possible ameliorative role of Thymus vulgaris (T. vulgaris) essential oil against Escherichia coli O157:H7 (E. coli O157:H7) deleterious effects in both blood and different tissues of rats by assessing the hematological, biochemical and immune-inflammatory parameters besides the histopathological alterations in the different organs. Forty male rats were randomly divided into four equal groups as follows: group I served as control, group II orally inoculated with E. coli O157:H7 at a dose of 1.0 × 109 cfu/ml, group III orally received 250 mg/kg BW T. vulgaris oil daily for 7 days and group IV orally inoculated with E. coli O157:H7 as the same dose of group II and orally received T. vulgaris oil as the same dose and duration of group III. Bacterial challenge in groups II and IV was once at the beginning of experiment and administration of oil began after 72 h from bacterial inoculation. At the end of the study, blood was sampled and complete blood picture, liver and kidney function alongside immunoglobulins and cytokines concentrations were estimated and tissues of large intestine (colon), liver and kidneys were collected for histopathological examinations. The results revealed that there was an increase of red blood cells count, hematocrit value and hemoglobin concentration besides white blood cells and thrombocytes counts and substantial increment of serum markers of hepatorenal damage such as the activities of transaminases and concentrations of bilirubin (total, direct and indirect), total proteins, albumin, creatinine and urea in E. coli O157:H7-challenged group. Also, there was a considerable increase in serum immunoglobulins M and G, interleukin 6 and 8 and tumor necrosis factor alpha as well as decreased serum alkaline phosphatase activity. Moreover, T. vulgaris oil could partially improve the hematological, biochemical and histopathological alterations induced by E. coli O157:H7 without any significant alterations in all measured parameters when used alone. The study concluded that the T. vulgaris oil relatively diminished the alterations in hematological parameters, hepatic and renal function markers and immune-inflammatory variables alongside the histopathological changes in different organs induced by E. coli O157:H7. The ameliorative effects of T. vulgaris oil are mediated through its anti-inflammatory, antioxidant and immunomodulatory activities.
Collapse
Affiliation(s)
- Hager Tarek H Ismail
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, 1 Alzeraa Street, Zagazig City, Postal Code 44511, Sharkia Province, Egypt.
| |
Collapse
|
18
|
Surfactant Protein D Influences Mortality During Abdominal Sepsis by Facilitating Escherichia coli Colonization in the Gut. Crit Care Explor 2022; 4:e0699. [PMID: 35620769 PMCID: PMC9119639 DOI: 10.1097/cce.0000000000000699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
19
|
Cuccato M, Scaglione FE, Centelleghe C, Divari S, Biolatti B, Pregel P, Cannizzo FT. Assessment of Antimicrobial Effects on Broiler Gut Barrier Through Histopathology and Immunohistochemistry of Tight-Junction Proteins. Front Vet Sci 2022; 9:830073. [PMID: 35425830 PMCID: PMC9002056 DOI: 10.3389/fvets.2022.830073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
In recent years, antimicrobial (AM) use in poultry farming has been attracting attention worldwide mainly due to AM resistance spreading. The role of AM prophylaxis in the modulation of gut microbiota, as well as of gut health, is still not clearly understood. Therefore, this study aimed to investigate the role of different prophylaxis protocols in the modulation of the gut barrier in broilers by applying a histopathological approach. Intestinal tissue samples were collected from a total of 240 male broilers (Ross 306), reared and treated with different AM protocols. Haematoxylin and Eosin (HE) staining and a multiple scoring system were used to evaluate the presence of lesions in ileum, cecum and colon of treated broilers. Moreover, immunohistochemistry (IHC) was performed to assess the expression of claudin-3 and ZO-1 proteins in intestinal tissues. The application of a semi-quantitative scoring system was used in IHC stained samples. HE results revealed that intestinal tissues were mainly characterized by epithelial detachment and fusion of the intestinal villi, but also by the presence of lymphocytic infiltrate in the mucosa and submucosa of AM-treated broilers. However, the IHC approach for the evaluation of claudin-3 and ZO-1 proteins showed that their expression was not affected by the different AM treatments. Nevertheless, the presence of intestinal lesions highlighted by histopathology suggests that AM treatments could harm the gut health of broilers, inducing an inflammatory response and consequent epithelial lesions. In order to clarify the role of AM treatments in the modulation of gut barrier in broilers, further studies are needed.
Collapse
Affiliation(s)
- Matteo Cuccato
- Department of Veterinary Science, University of Turin, Turin, Italy
| | | | - Cinzia Centelleghe
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Sara Divari
- Department of Veterinary Science, University of Turin, Turin, Italy
- *Correspondence: Sara Divari
| | | | - Paola Pregel
- Department of Veterinary Science, University of Turin, Turin, Italy
| | | |
Collapse
|
20
|
Park NY, Koh A. From the Dish to the Real World: Modeling Interactions between the Gut and Microorganisms in Gut Organoids by Tailoring the Gut Milieu. Int J Stem Cells 2022; 15:70-84. [PMID: 35220293 PMCID: PMC8889331 DOI: 10.15283/ijsc21243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/11/2022] Open
Abstract
The advent of human intestinal organoid systems has revolutionized the way we understand the interactions between the human gut and microorganisms given the host tropism of human microorganisms. The gut microorganisms have regionality (i.e., small versus large intestine) and the expression of various virulence factors in pathogens is influenced by the gut milieu. However, the culture conditions, optimized for human intestinal organoids, often do not fully support the proliferation and functionality of gut microorganisms. In addition, the regional identity of human intestinal organoids has not been considered to study specific microorganisms with regional preference. In this review we provide an overview of current efforts to understand the role of microorganisms in human intestinal organoids. Specifically, we will emphasize the importance of matching the regional preference of microorganisms in the gut and tailoring the appropriate luminal environmental conditions (i.e., oxygen, pH, and biochemical levels) for modeling real interactions between the gut and the microorganisms with human intestinal organoids.
Collapse
Affiliation(s)
- Na-Young Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| |
Collapse
|
21
|
Painong San, a Traditional Chinese Compound Herbal Medicine, Restores Colon Barrier Function on DSS-Induced Colitis in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2810915. [PMID: 34966434 PMCID: PMC8712168 DOI: 10.1155/2021/2810915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/10/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022]
Abstract
Objective The intestinal barrier decreases in colitis and restores the integrity of the mucosal barriers that could be used for the treatment of colitis. Painong San (PNS), a traditional Chinese compound herbal medicine originally recorded in “Jingui Yaolve” by Zhongjing Zhang in the Later Han Dynasty, is often used in China and Japan to treat various purulent diseases including intestinal carbuncle. This study was to investigate the effect of PNS on mucosal barrier function in mice with DSS-induced colitis and its related mechanisms. Methods BALB/C mice were given 3% DSS to induce colitis. The body weight and stool status of the mice were recorded daily, and the histopathological changes of the colon were observed after execution. The permeability of the intestinal mucosa was measured by fluorescein isothiocyanate-dextran 4000, the change of intestinal microbiota was measured by 16S rDNA, and the tight junction-related proteins and Muc-2 were investigated by immunohistochemical or immunofluorescence. The possible signaling pathways were detected by western blot. Results Compared with the control group, the composition of the microbiota in the PNS group was close to that of the normal group, the number of goblet cells was improved, and the mucosal permeability was significantly reduced. PNS could upregulate the expression of tight junction-related proteins (ZO-1, claudin-1, and occludin) and Muc-2, and at the same time, regulate the Notch pathway. Conclusion PNS could effectively improve the mucosal barrier function through multiple ways, including restoring the balance of intestine flora, enhancement of the mucous layer barrier, and mechanical barrier function. These protective effects may relate to inhibiting the Notch signaling pathway activated by DSS.
Collapse
|
22
|
Serek P, Oleksy-Wawrzyniak M. The Effect of Bacterial Infections, Probiotics and Zonulin on Intestinal Barrier Integrity. Int J Mol Sci 2021; 22:11359. [PMID: 34768787 PMCID: PMC8583036 DOI: 10.3390/ijms222111359] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal barrier plays an extremely important role in maintaining the immune homeostasis of the gut and the entire body. It is made up of an intricate system of cells, mucus and intestinal microbiota. A complex system of proteins allows the selective permeability of elements that are safe and necessary for the proper nutrition of the body. Disturbances in the tightness of this barrier result in the penetration of toxins and other harmful antigens into the system. Such events lead to various digestive tract dysfunctions, systemic infections, food intolerances and autoimmune diseases. Pathogenic and probiotic bacteria, and the compounds they secrete, undoubtedly affect the properties of the intestinal barrier. The discovery of zonulin, a protein with tight junction regulatory activity in the epithelia, sheds new light on the understanding of the role of the gut barrier in promoting health, as well as the formation of diseases. Coincidentally, there is an increasing number of reports on treatment methods that target gut microbiota, which suggests that the prevention of gut-barrier defects may be a viable approach for improving the condition of COVID-19 patients. Various bacteria-intestinal barrier interactions are the subject of this review, aiming to show the current state of knowledge on this topic and its potential therapeutic applications.
Collapse
Affiliation(s)
- Paweł Serek
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Oleksy-Wawrzyniak
- Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
23
|
Ma J, Piao X, Shang Q, Long S, Liu S, Mahfuz S. Mixed organic acids as an alternative to antibiotics improve serum biochemical parameters and intestinal health of weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:737-749. [PMID: 34466678 PMCID: PMC8379140 DOI: 10.1016/j.aninu.2020.11.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/09/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
The primary aim of this experiment was to critically explore the relationship between the different levels of mixed organic acids (MOA) and growth performance, serum antioxidant status and intestinal health of weaned piglets, as well as to investigate the potential possibility of MOA alternative to antibiotics growth promoters (AGP). A total of 180 healthy piglets (Duroc × [Landrace × Yorkshire]; weighing 7.81 ± 1.51 kg each, weaned at d 28) were randomly divided into 5 treatments: 1) basal diet (CON); 2) CON + chlorinomycin (75 mg/kg) + virginiamycin (15 mg/kg) + guitaromycin (50 mg/kg) (AGP); 3) CON + MOA (3,000 mg/kg) (OA1); 4) CON + MOA (5,000 mg/kg) (OA2); 5) CON + MOA (7,000 mg/kg) (OA3). This study design included 6 replicates per treatment with 6 piglets per pen (barrow:gilt = 1:1) and the experiment was separated into phase 1 (d 1 to 14) and phase 2 (d 15 to 28). In phases 1, 2 and overall, compared with the CON, the feed conversion ratio (FCR) was reduced (P < 0.01) and the average daily gain (ADG) was increased (P < 0.05) in piglets supplemented with AGP, OA1 and OA2. The concentration of serum immunoglobulins G (IgG) was improved (P < 0.05) in piglets supplemented with OA2 in phase 2. In the jejunum and ileum, the villus height:crypt depth ratio was significantly increased (P < 0.01) in piglets fed AGP and OA1. The mRNA expression level of claudin-1 and zonula occludens-1 (ZO-1) (P < 0.01) was up-regulated in piglets supplemented with OA1 and OA2. The piglets fed AGP, OA1 and OA2 showed an increase (P < 0.05) in the content of acetate acid and total volatile fatty acids (TVFA) in the cecum, and butyric acid and TVFA in the colon compared with CON. Also, OA1 lowered (P < 0.05) the content of Lachnospiraceae in piglets. These results demonstrated that MOA at 3,000 or 5,000 mg/kg could be an alternative to antibiotics due to the positive effects on performance, immune parameters, and intestinal health of weaned piglets. However, from the results of the quadratic fitting curve, it is inferred that MOA at a dose of 4,000 mg/kg may produce a better effect.
Collapse
Affiliation(s)
- Jiayu Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qinghui Shang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shenfei Long
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Sujie Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shad Mahfuz
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
24
|
Xue Y, Zhu MJ. Unraveling enterohemorrhagic Escherichia coli infection: the promising role of dietary compounds and probiotics in bacterial elimination and host innate immunity boosting. Crit Rev Food Sci Nutr 2021; 63:1551-1563. [PMID: 34404306 DOI: 10.1080/10408398.2021.1965538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The innate immune system has developed sophisticated strategies to defense against infections. Host cells utilize the recognition machineries such as toll-like receptors and nucleotide binding and oligomerization domain-like receptors to identify the pathogens and alert immune system. However, some pathogens have developed tactics to evade host defenses, including manipulation of host inflammatory response, interference with cell death pathway, and highjack of phagocytosis signaling for a better survival and colonization in host. Enterohemorrhagic Escherichia coli (EHEC) is a notorious foodborne pathogen that causes severe tissue damages and gastrointestinal diseases, which has been reported to disturb host immune responses. Diverse bioactive compounds such as flavonoids, phenolic acids, alkaloids, saccharides, and terpenoids derived from food varieties and probiotics have been discovered and investigated for their capability of combating bacterial infections. Some of them serve as novel antimicrobial agents and act as immune boosters that harness host immune system. In this review, we will discuss how EHEC, specifically E. coli O157:H7, hijacks the host immune system and interferes with host signaling pathway; and highlight the promising role of food-derived bioactive compounds and probiotics in harnessing host innate immunity and eliminating E. coli O157:H7 infection with multiple strategies.
Collapse
Affiliation(s)
- Yansong Xue
- Key Laboratory of Functional Dairy, Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, USA
| |
Collapse
|
25
|
Young RB, Marcelino VR, Chonwerawong M, Gulliver EL, Forster SC. Key Technologies for Progressing Discovery of Microbiome-Based Medicines. Front Microbiol 2021; 12:685935. [PMID: 34239510 PMCID: PMC8258393 DOI: 10.3389/fmicb.2021.685935] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/25/2021] [Indexed: 12/22/2022] Open
Abstract
A growing number of experimental and computational approaches are illuminating the “microbial dark matter” and uncovering the integral role of commensal microbes in human health. Through this work, it is now clear that the human microbiome presents great potential as a therapeutic target for a plethora of diseases, including inflammatory bowel disease, diabetes and obesity. The development of more efficacious and targeted treatments relies on identification of causal links between the microbiome and disease; with future progress dependent on effective links between state-of-the-art sequencing approaches, computational analyses and experimental assays. We argue determining causation is essential, which can be attained by generating hypotheses using multi-omic functional analyses and validating these hypotheses in complex, biologically relevant experimental models. In this review we discuss existing analysis and validation methods, and propose best-practice approaches required to enable the next phase of microbiome research.
Collapse
Affiliation(s)
- Remy B Young
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Vanessa R Marcelino
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Emily L Gulliver
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
26
|
Flowers LJ, Hu S, Shrestha A, Martinot AJ, Leong JM, Osburne MS. Citrobacter rodentium Lysogenized with a Shiga Toxin-Producing Phage: A Murine Model for Shiga Toxin-Producing E. coli Infection. Methods Mol Biol 2021; 2291:381-397. [PMID: 33704765 DOI: 10.1007/978-1-0716-1339-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Shiga toxin-producing E. coli (STEC) is a common foodborne pathogen in developed countries. STEC generates "attaching and effacing" (AE) lesions on colonic epithelium, characterized by effacement of microvilli and the formation of actin "pedestals" beneath intimately attached bacteria. In addition, STEC are lysogenized with a phage that, upon induction, can produce potent Shiga toxins (Stx), potentially leading to both hemorrhagic colitis and hemolytic uremic syndrome. Investigation of the pathogenesis of this disease has been challenging because STEC does not readily colonize conventional mice.Citrobacter rodentium (CR) is a related mouse pathogen that also generates AE lesions. Whereas CR does not produce Stx, a murine model for STEC utilizes CR lysogenized with an E. coli-derived Stx phage, generating CR(Φstx), which both colonizes conventional mice and readily gives rise to systemic disease. We present here key methods for the use of CR(Φstx) infection as a highly predictable murine model for infection and disease by STEC. Importantly, we detail CR(Φstx) inoculation by feeding, determination of pathogen colonization, production of phage and toxin, and assessment of intestinal and renal pathology. These methods provide a framework for studying STEC-mediated systemic disease that may aid in the development of efficacious therapeutics.
Collapse
Affiliation(s)
- Laurice J Flowers
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA.,Tufts University Graduate School in Biomedical Sciences, Boston, MA, USA.,Department of Dermatology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shenglan Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA.,Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding, Guangzhou, China
| | - Anishma Shrestha
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Amanda J Martinot
- Department of Infectious Diseases and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Marcia S Osburne
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
27
|
Tight Junctions as a Key for Pathogens Invasion in Intestinal Epithelial Cells. Int J Mol Sci 2021; 22:ijms22052506. [PMID: 33801524 PMCID: PMC7958858 DOI: 10.3390/ijms22052506] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junctions play a major role in maintaining the integrity and impermeability of the intestinal barrier. As such, they act as an ideal target for pathogens to promote their translocation through the intestinal mucosa and invade their host. Different strategies are used by pathogens, aimed at directly destabilizing the junctional network or modulating the different signaling pathways involved in the modulation of these junctions. After a brief presentation of the organization and modulation of tight junctions, we provide the state of the art of the molecular mechanisms leading to permeability breakdown of the gut barrier as a consequence of tight junctions’ attack by pathogens, including bacteria, viruses, fungi, and parasites.
Collapse
|
28
|
Guo Y, Tu YH, Wu X, Ji S, Shen JL, Wu HM, Fei GH. ResolvinD1 Protects the Airway Barrier Against Injury Induced by Influenza A Virus Through the Nrf2 Pathway. Front Cell Infect Microbiol 2021; 10:616475. [PMID: 33643931 PMCID: PMC7907644 DOI: 10.3389/fcimb.2020.616475] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/16/2020] [Indexed: 11/13/2022] Open
Abstract
Airway barrier damage and excessive inflammation induced by influenza A virus (IAV) are associated with disease progression and prognosis. ResolvinD1 (RvD1) is a promising lipid mediator with critical protection against infection in the lung. However, whether RvD1 protects against IAV-induced injury and the underlying mechanisms remain elusive. In this study, primary normal human bronchial epithelial (pNHBE) cells were isolated and co-cultured with IAV and/or RvD1. Then, the expressions of E-cadherin, Zonula occludins-1, inflammatory mediators and proteins in Nrf2-dependent pathway were detected. To further explore the mechanisms, Nrf2 short hairpin RNA (Nrf2 shRNA) was applied in pNHBE cells. Furthermore, mice were infected with IAV, and were subsequently treated with RvD1. We found that IAV downregulated expressions of E-cadherin, Zonula occludins-1, Nrf2 and HO-1, upregulated the phosphorylation of NF κ B p65 and IKBα, levels of IL-8 and TNF-α, as well as ROS production. RvD1 reversed these damaging effects induced by IAV. However, when Nrf2 expression was suppressed with shRNA in pNHBE cells, the protective effects of RvD1 on IAV-induced injury were inhibited. In vivo studies further demonstrated that RvD1 could alleviate barrier protein breakdown and reduce airway inflammatory reactions. Collectively, the study demonstrated that RvD1 could play dual beneficial roles in protecting airway epithelium barrier function and reducing inflammation via the Nrf2 pathway, which may provide a better treatment option for influenza A virus infection.
Collapse
Affiliation(s)
- Yan Guo
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - You-Hui Tu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xu Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuang Ji
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ji-Long Shen
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Hui-Mei Wu
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Chanez-Paredes SD, Abtahi S, Kuo WT, Turner JR. Differentiating Between Tight Junction-Dependent and Tight Junction-Independent Intestinal Barrier Loss In Vivo. Methods Mol Biol 2021; 2367:249-271. [PMID: 33830456 PMCID: PMC8249353 DOI: 10.1007/7651_2021_389] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The intestinal barrier is an essential component of innate host defense. The single layer of epithelial cells that line the intestine must balance barrier function with both active, transcellular and diffusive, paracellular transport. Tight junctions, which link adjacent cells, form a selectively permeable seal that defines both paracellular transport and barrier properties. Molecules can cross tight junctions by either of two distinct routes, termed pore and the leak pathways, that differ in capacity, charge-selectivity, size-selectivity, and responses to physiological and pathophysiological stimuli. A third intestinal permeability route, the unrestricted pathway, reflects loss of the epithelial barrier, as occurs with mucosal damage, is independent of paracellular and transcellular pathways, and is neither charge- nor size-selective.The most commonly used approach for measuring intestinal permeability in vivo involves gavage of FITC-4 kDa dextran and analysis of the quantity recovered in serum. Unfortunately, this method cannot distinguish between leak and unrestricted pathways, as 4 kDa dextran can cross both. Moreover, 4 kDa dextran is too large to cross the pore pathway and, therefore, provides no information regarding this paracellular flux route. Here we describe a multiplex method that allows simultaneous, independent analysis of each pathway.
Collapse
Affiliation(s)
- Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wei-Ting Kuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Shi J, Xie Q, Yue Y, Chen Q, Zhao L, Evivie SE, Li B, Huo G. Gut microbiota modulation and anti-inflammatory properties of mixed lactobacilli in dextran sodium sulfate-induced colitis in mice. Food Funct 2021; 12:5130-5143. [PMID: 33973599 DOI: 10.1039/d1fo00317h] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Correlations between gut microbiota activities and inflammatory bowel disease (IBD) treatment are gaining research interest. In our previous study, Lactobacillus acidophilus KLDS 1.0901, Lactobacillus helveticus KLDS 1.8701, and Lactobacillus plantarum KLDS 1.0318 showed antibacterial, antioxidant, and immunomodulatory activities. In the current study, we evaluated the effects of three tested strains and their mixture on dextran sulfate sodium (DSS)-induced colitis in C57BL/6J mice. The three tested strains and their mixture significantly decreased the disease activity index (DAI), colon shortening, and myeloperoxidase (MPO) activity. Additionally, the three tested strains and their mixture improved the histological damage, increased the colonic mucous layer integrity, and exhibited lower levels of prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), while up-regulating colonic anti-inflammatory cytokine IL-10 levels, tight junction proteins (E-cadherin, zonulae occludens (ZO)-1, occludin and claudin-1) and mucin (MUC1 and MUC2) mRNA expressions to some extent. In addition, mixed lactobacilli showed better anti-inflammatory effects than single-strain treatment. Our study further revealed that mixed lactobacilli increased bacterial diversity and improved gut microbiota composition, increasing short-chain fatty acid (SCFA) production. These results indicated that mixed lactobacilli supplementation could attenuate DSS-induced colitis by modulating the gut microbiota and repairing the intestinal barrier, which provided a scientific basis for its clinical application in the future.
Collapse
Affiliation(s)
- Jialu Shi
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Qinggang Xie
- Heilongjiang Feihe Dairy Co., LTD, Qiqihaer 164800, China
| | - Yingxue Yue
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Qingxue Chen
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Lina Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Smith Etareric Evivie
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China. and Department of Food Science and Human Nutrition, Faculty of Agriculture, University of Benin, Benin City 300001, Nigeria and Department of Animal Science, Faculty of Agriculture, University of Benin, Benin City 300001, Nigeria
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. and Food College, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
31
|
Hua Y, Wu J, Fu M, Liu J, Li X, Zhang B, Zhao W, Wan C. Enterohemorrhagic Escherichia coli Effector Protein EspF Interacts With Host Protein ANXA6 and Triggers Myosin Light Chain Kinase (MLCK)-Dependent Tight Junction Dysregulation. Front Cell Dev Biol 2020; 8:613061. [PMID: 33425920 PMCID: PMC7785878 DOI: 10.3389/fcell.2020.613061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/02/2020] [Indexed: 01/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is an important foodborne pathogen that can cause bloody diarrhea and hemolytic uremic syndrome (HUS) in humans. EspF is one of the best-characterized effector proteins secreted from the type three secretion system to hijack host cell functions. However, the crucial pathogen-host interactions and the basis for the intestinal barrier disruption during infections remain elusive. Our previous study screened and verified the interaction between host protein ANXA6 and EspF protein. Here, by fluorescence resonance energy transfer (FRET) and co-immunoprecipitation (CO-IP), we verified that EspF interacts with ANXA6 through its C-terminal domain. Furthermore, we found that both the constitutive expression of EspF or ANXA6 and the co-expression of EspF-ANXA6 could decrease the levels of tight junction (TJ) proteins ZO-1 and occludin, and disrupt the distribution of ZO-1. Moreover, we showed that EspF-ANXA6 activated myosin light chain kinase (MLCK), induced the phosphorylation of myosin light chain (MLC) and PKCα, and down-regulated the expression level of Calmodulin protein. Collectively, this study revealed a novel interaction between the host protein (ANXA6) and EspF. The binding of EspF to ANXA6 may perturb TJs in an MLCK-MLC-dependent manner, and thus may be involved in EHEC pathogenic function.
Collapse
Affiliation(s)
- Ying Hua
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Jiali Wu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Muqing Fu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinyue Liu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoxia Li
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Wei Zhao
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Chengsong Wan
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| |
Collapse
|
32
|
Low-Protein Diet Supplemented with Medium-Chain Fatty Acid Glycerides Improves the Growth Performance and Intestinal Function in Post-Weaning Piglets. Animals (Basel) 2020; 10:ani10101852. [PMID: 33053685 PMCID: PMC7601791 DOI: 10.3390/ani10101852] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary After weaning, piglets cannot absorb protein well and cannot get enough energy from the diet due to intestinal dysplasia. Medium-chain fatty acids are very effective in providing energy for piglets and may protect the integrity of the intestinal barrier to improve the healthy development of piglets. Therefore, we speculate that medium chain fatty acid glycerides can promote the growth of weaned piglets in a low protein diet. The present study examined the effects of medium-chain fatty acid glycerides on the growth performance, intestinal barrier function and inflammatory response of weaned piglets. These findings provide a new prospect for the application of medium-chain fatty acid triglycerides in piglets. Abstract Medium-chain fatty acid glycerides have been shown to provide energy for rapid oxidation in the body. The study was conducted to investigate the effects of dietary supplementation with medium-chain fatty acid glyceride on the growth performance and intestinal health of weaned piglets fed with a low-protein diet. Nighty healthy weaned piglets were randomly divided into five treatments: NP (Normal protein treatment, normal-protein diet no antibiotics included); NC (Negative control, low-protein diet no antibiotics included); PC (Positive control, low-protein diet +75 mg/kg quinocetone, 20 mg/kg virginiamycin and 50 mg/kg aureomycin); MCT (tricaprylin + tricaprin treatment, low-protein diet + tricaprylin + tricaprin); GML (glycerol monolaurate treatment, low-protein diet + glycerol monolaurate). The results showed that the average daily feed intake (ADFI) of the MCT treatment was significantly higher than that of the NP, NC treatments (p < 0.05). In the jejunum, the villus height of the GML treatment was significantly lower than that of the PC treatment (p < 0.05), and the number of goblet cells in the GML treatment was higher than that in the NC treatment (p < 0.05). Compared with the NC treatment, the MCT treatment significantly increased the level of claudin-1, Zonula occludens-1(ZO-1), while the GML treatment significantly increased the level of claudin-1, occludin, ZO-1 (p < 0.05). In the ileum, the level of ZO-1 in the GML treatment was significantly higher than that in the NP, NC, PC treatments (p < 0.05). Compared with the NC treatment, the GML treatment significantly increased the level of Secretory immunoglobulin A (SIgA) in the ileum and serum, while the MCT treatment significantly increased the level of SIgA and decreased the level of interleukin-6 (IL-6) in the ileum (p < 0.05). These results showed that the addition of medium-chain fatty acid glycerides to a low-protein diet could improve the growth performance and intestinal functional barrier of weaned piglets and also improve the immune function of weaned piglets.
Collapse
|
33
|
Chitosan-chelated zinc modulates cecal microbiota and attenuates inflammatory response in weaned rats challenged with Escherichia coli. J Microbiol 2020; 58:780-792. [PMID: 32870484 DOI: 10.1007/s12275-020-0056-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/08/2023]
Abstract
Escherichia coli (E. coli) infection is very common among young growing animals, and zinc supplementation is often used to alleviate inflammation induced by this disease. Therefore, the objective of this study was to evaluate whether chitosan-chelated zinc (CS-Zn) supplementation could attenuate gut injury induced by E. coli challenge and to explore how CS-Zn modulates cecal microbiota and alleviates intestinal inflammation in weaned rats challenged with E. coli. 36 weaned rats (55.65 ± 2.18 g of BW, n = 12) were divided into three treatment groups consisting of unchallenged rats fed a basal diet (Control) and two groups of rats challenged with E. coli and fed a basal diet or a diet containing 640 mg/kg CS-Zn (E. coli + CS-Zn, containing 50 mg/kg Zn) for a 14-day experiment. On days 10 to 12, each rat was given 4 ml of E. coli solution with a total bacteria count of 1010 CFU by oral gavage daily or normal saline of equal dosage. CS-Zn supplementation mitigated intestinal morphology impairment (e.g. higher crypt depth and lower macroscopic damage index) induced by E. coli challenge (P < 0.05), and alleviated the increase of Myeloperoxidase (MPO) activity after E. coli challenge (P < 0.05). 16S rRNA sequencing analyses revealed that E. coli challenge significantly increased the abundance of Verrucomicrobia and E. coli (P < 0.05). However, CS-Zn supplementation increased the abundance of Lactobacillus and decreased the relative abundance of Proteobacteria, Desulfovibrio and E. coli (P < 0.05). The concentrations of butyrate in the cecal digesta, which decreased due to the challenge, were higher in the E. coli + CS-Zn group (P < 0.05). In addition, CS-Zn supplementation significantly prevented the elevation of pro-inflammatory cytokines IL-6 concentration and up-regulated the level of anti-inflammatory cytokines IL-10 in cecal mucosa induced by E. coli infection (P < 0.05). In conclusion, these results indicate that CS-Zn produces beneficial effects in alleviating gut mucosal injury of E. coli challenged rats by enhancing the intestinal morphology and modulating cecal bacterial composition, as well as attenuating inflammatory response.
Collapse
|
34
|
Leon-Coria A, Kumar M, Workentine M, Moreau F, Surette M, Chadee K. Muc2 Mucin and Nonmucin Microbiota Confer Distinct Innate Host Defense in Disease Susceptibility and Colonic Injury. Cell Mol Gastroenterol Hepatol 2020; 11:77-98. [PMID: 32659381 PMCID: PMC7596264 DOI: 10.1016/j.jcmgh.2020.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Alterations in intestinal MUC2 mucin and microbial diversity are closely linked with important intestinal pathologies; however, their impact on each other and on intestinal pathogenesis has been vaguely characterized. Therefore, it was of interest in this study to delineate distinct and cooperative function of commensal microbiota and the Muc2 mucus barrier in maintaining intestinal epithelial barrier function. METHODS Muc2 mucin deficient (Muc2-/-) and sufficient (Muc2+/+) littermates were used as a model for assessing the role of Muc2. To quantify the role of the microbiota in disease pathogenesis, Muc2+/+ and Muc2-/- littermates were treated with a cocktail of antibiotics that reduced indigenous bacteria, and then fecal transplanted with littermate stool and susceptibility to dextran sulphate sodium (DSS) quantified. RESULTS Although, Muc2+/+ and Muc2-/- littermates share similar phyla distribution as evidenced by 16S sequencing they maintain their distinctive gastrointestinal phenotypes. Basally, Muc2-/- showed low-grade colonic inflammation with high populations of inflammatory and tolerogenic immune cells that became comparable to Muc2+/+ littermates following antibiotic treatment. Antibiotics treatment rendered Muc2+/+ but not Muc2-/- littermates highly susceptibility to DSS-induced colitis that was ILC3 dependent. Muc2-/- microbiota was colitogenic to Muc2+/+ as it worsened DSS-induced colitis. Microbiota dependent inflammation was confirmed by bone-marrow chimera studies, as Muc2-/- receiving Muc2+/+ bone marrow showed no difference in their susceptibility toward DSS induced colitis. Muc2-/- microbiota exhibited presence of characteristic OTUs of specific bacterial populations that were transferrable to Muc2+/+ littermates. CONCLUSIONS These results highlight a distinct role for Muc2 mucin in maintenance of healthy microbiota critical in shaping innate host defenses to promote intestinal homeostasis.
Collapse
Affiliation(s)
- Aralia Leon-Coria
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Manish Kumar
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Matthew Workentine
- Department of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Michael Surette
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada.
| |
Collapse
|
35
|
The development of intestinal dysbiosis in anemic preterm infants. J Perinatol 2020; 40:1066-1074. [PMID: 31992818 PMCID: PMC7319903 DOI: 10.1038/s41372-020-0599-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Anemia and Proteobacteria-dominant intestinal dysbiosis in very low birth weight (VLBW) infants have been linked to necrotizing enterocolitis, a severe gut inflammatory disease. We hypothesize that anemia of prematurity is related to the development of intestinal dysbiosis. STUDY DESIGN Three hundred and forty-two weekly stool samples collected prospectively from 80 VLBW infants were analyzed for bacterial microbiomes (with 16S rRNA). Linear mixed-effects model was used to determine the relationships between the onsets of anemia and intestinal dysbiosis. RESULTS Hematocrit was associated with intestinal microbiomes, with lower Hct occurring with increased Proteobacteria and decreased Firmicutes. Infants with a hematocrit <30% had intestinal microbiomes that diverged toward Proteobacteria dominance and low diversity after the first postnatal month. The microbiome changes were also related to the severity of anemia. CONCLUSIONS This finding supports a potential microbiological explanation for anemia as a risk factor for intestinal dysbiosis in preterm infants.
Collapse
|
36
|
Immune-Microbiota Interplay and Colonization Resistance in Infection. Mol Cell 2020; 78:597-613. [PMID: 32208169 DOI: 10.1016/j.molcel.2020.03.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/12/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
|
37
|
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: 0.8] [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
|
38
|
Detection and isolation of protein susceptible indigenous bacteria affected by dietary milk-casein, albumen and soy-protein in the caecum of ICR mice. Int J Biol Macromol 2020; 144:813-820. [DOI: 10.1016/j.ijbiomac.2019.09.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/16/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023]
|
39
|
Zhang Y, Chen S, Zong X, Wang C, Shi C, Wang F, Wang Y, Lu Z. Peptides derived from fermented soybean meal suppresses intestinal inflammation and enhances epithelial barrier function in piglets. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1705766] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Yu Zhang
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shan Chen
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xin Zong
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Cheng Wang
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Changyou Shi
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fengqin Wang
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| | - Zeqing Lu
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture (East China), Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Institute of Feed Science, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
40
|
Zhang M, Kou J, Wu Y, Wang M, Zhou X, Yang Y, Wu Z. Dietary genistein supplementation improves intestinal mucosal barrier function in Escherichia coli O78-challenged broilers. J Nutr Biochem 2019; 77:108267. [PMID: 32000135 DOI: 10.1016/j.jnutbio.2019.108267] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/28/2019] [Accepted: 11/03/2019] [Indexed: 12/23/2022]
Abstract
Genistein has multiple biological activities in both humans and animals. However, a protective effect of genistein on Escherichia coli (E. coli)-induced intestinal mucosal barrier dysfunction remains unknown. In the present study, a total of 288 1-day-old male Arbor Acre broilers fed a corn-soybean basal diet unsupplemented or supplemented with 20 mg genistein/kg diet were subjected to E. coli serotype O78 (108 cfu per bird) infection or equal volume of sodium chloride at 19 days of age. Sera and tissue samples were collected 2 days after E. coli infection. Growth performance, index of immune-related organs, intestinal barrier permeability, protein level of inflammatory cytokines, sIgA, tight junction protein, and mRNA level of apoptotic genes in jejunum were determined. Mortality rate at 7 days post infection was recorded. The results showed that E. coli challenge led to a reduced average daily gain, a decreased thymus index, and bursal index in broilers, an increase of fluorescein isothiocyanate (FITC)-dextran in serum, and a decreased sIgA in jejunum. These effects were abrogated by genistein administration. Western blot results showed that E. coli infection led to increased protein level of claudin-1 and zonula occludens (ZO)-1, which was largely abolished by genistein. Moreover, E. coli infection resulted increased protein level of TNF-α and IL-6, enhanced mRNA level of Bax and caspase-3, as well as decreased mRNA level of Bcl-2 were abrogated by genistein in jujunum of broilers. In conclusion, the results indicate that genistein supplementation improves intestinal mucosal barrier function which is associated with a regulatory effect on tight junction proteins, sIgA, apoptosis, and secretion of inflammatory cytokines in jejunum of E. coli-challenged broilers.
Collapse
Affiliation(s)
- Ming Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Jiao Kou
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Mengmeng Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Xiumin Zhou
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
41
|
Enterohemorrhagic Escherichia coli infection inhibits colonic thiamin pyrophosphate uptake via transcriptional mechanism. PLoS One 2019; 14:e0224234. [PMID: 31639155 PMCID: PMC6804999 DOI: 10.1371/journal.pone.0224234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/08/2019] [Indexed: 01/19/2023] Open
Abstract
Colonocytes possess a specific carrier-mediated uptake process for the microbiota-generated thiamin (vitamin B1) pyrophosphate (TPP) that involves the TPP transporter (TPPT; product of the SLC44A4 gene). Little is known about the effect of exogenous factors (including enteric pathogens) on the colonic TPP uptake process. Our aim in this study was to investigate the effect of Enterohemorrhagic Escherichia coli (EHEC) infection on colonic uptake of TPP. We used human-derived colonic epithelial NCM460 cells and mice in our investigation. The results showed that infecting NCM460 cells with live EHEC (but not with heat-killed EHEC, EHEC culture supernatant, or with non-pathogenic E. Coli) to lead to a significant inhibition in carrier-mediated TPP uptake, as well as in level of expression of the TPPT protein and mRNA. Similarly, infecting mice with EHEC led to a significant inhibition in colonic TPP uptake and in level of expression of TPPT protein and mRNA. The inhibitory effect of EHEC on TPP uptake by NCM460 was found to be associated with reduction in the rate of transcription of the SLC44A4 gene as indicated by the significant reduction in the activity of the SLC44A4 promoter transfected into EHEC infected cells. The latter was also associated with a marked reduction in the level of expression of the transcription factors CREB-1 and ELF3, which are known to drive the activity of the SLC44A4 promoter. Finally, blocking the ERK1/2 and NF-kB signaling pathways in NCM460 cells significantly reversed the level of EHEC inhibition in TPP uptake and TPPT expression. Collectively, these findings show, for the first time, that EHEC infection significantly inhibit colonic uptake of TPP, and that this effect appears to be exerted at the level of SLC44A4 transcription and involves the ERK1/2 and NF-kB signaling pathways.
Collapse
|
42
|
Haiwen Z, Rui H, Bingxi Z, Qingfeng G, Beibei W, Jifeng Z, Xuemei W, Kebang W. Cathelicidin- derived PR39 protects enterohemorrhagic Escherichia coli O157:H7 challenged mice by improving epithelial function and balancing the microbiota in the intestine. Sci Rep 2019; 9:9456. [PMID: 31263234 PMCID: PMC6603261 DOI: 10.1038/s41598-019-45913-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
The zoonotic enterohaemorrhagic Escherichia coli (EHEC) O157:H7 can disrupt intestinal epithelial barrier function and in turn leading to serious intestinal and systemic disease. PR39 could effectively inhibit the growth of Gram-negative bacteria, but there is little knowledge of its effects on intestinal barrier function and the microbiota in E. coli-challenged mice. In this study, an intestinal disease caused by EHEC O157:H7 was established, to analyze the effect of PR39 on EHEC O157:H7 induced intestinal epithelial barrier injury and disorder. Interestingly, PR39 attenuated EHEC O157:H7-induced systemic symptoms and significantly decreased mortality and the degree of E. coli shedding in faeces. Furthermore, the infiltration index of macrophages and neutrophils in intestine of the PR39 treatment group were obviously attenuated, along with the level of apoptosis. PR39 treatment group had distinctly improved tight junction associated proteins’ expression after EHEC O157:H7 caused injury. Additionally, the sequencing analysis of cecum microbiota showed that PR39 altered the abnormal increase in Bacteroides caused by EHEC O157:H7 and promoted the growth of probiotics such as Lactobacillus. In conclusion, cathelicidin-derived PR39 could effectively improve EHEC O157:H7-induced epithelial barrier injury, and dysfunction of immune and microbiota homeostasis in the intestinal tract, indicating that PR39 could be an excellent potential drug for zoonotic EHEC O157:H7-related intestinal disease.
Collapse
Affiliation(s)
- Zhang Haiwen
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China
| | - Hua Rui
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Zhang Bingxi
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Guan Qingfeng
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China
| | - Wang Beibei
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China.
| | - Zeng Jifeng
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China
| | - Wang Xuemei
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China
| | - Wu Kebang
- Tropical animal breeding and nutrition laboratory, Hainan University, Haikou, Hainan, 570228, People's Republic of China. .,Key Laboratory of Tropical Biological Resources of Ministry of Education, Haikou, Hainan, 570228, People's Republic of China.
| |
Collapse
|
43
|
Stromberg ZR, Van Goor A, Redweik GAJ, Wymore Brand MJ, Wannemuehler MJ, Mellata M. Pathogenic and non-pathogenic Escherichia coli colonization and host inflammatory response in a defined microbiota mouse model. Dis Model Mech 2018; 11:dmm035063. [PMID: 30275104 PMCID: PMC6262807 DOI: 10.1242/dmm.035063] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022] Open
Abstract
Most Escherichia coli strains in the human intestine are harmless. However, enterohemorrhagic Ecoli (EHEC) is a foodborne pathogen that causes intestinal disease in humans. Conventionally reared (CONV) mice are inconsistent models for human infections with EHEC because they are often resistant to Ecoli colonization, in part due to their gastrointestinal (GI) microbiota. Although antibiotic manipulation of the mouse microbiota has been a common means to overcome colonization resistance, these models have limitations. Currently, there are no licensed treatments for clinical EHEC infections and, thus, new tools to study EHEC colonization need to be developed. Here, we used a defined microbiota mouse model, consisting of the altered Schaedler flora (ASF), to characterize intestinal colonization and compare host responses following colonization with EHEC strain 278F2 or non-pathogenic Ecoli strain MG1655. Significantly higher (P<0.05) levels of both strains were found in feces and cecal and colonic contents of C3H/HeN ASF compared to C3H/HeN CONV mice. GI inflammation was significantly elevated (P<0.05) in the cecum of EHEC 278F2-colonized compared to E. coli MG1655-colonized C3H/HeN ASF mice. In addition, EHEC 278F2 differentially modulated inflammatory-associated genes in colonic tissue of C3H/HeN ASF mice compared to E. coli MG1655-colonized mice. This approach allowed for prolonged colonization of the murine GI tract by pathogenic and non-pathogenic Ecoli strains, and for evaluation of host inflammatory processes. Overall, this system can be used as a powerful tool for future studies to assess therapeutics, microbe-microbe interactions, and strategies for preventing EHEC infections.
Collapse
Affiliation(s)
- Zachary R Stromberg
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Angelica Van Goor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Graham A J Redweik
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Meghan J Wymore Brand
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
44
|
Roxas JL, Monasky RC, Roxas BAP, Agellon AB, Mansoor A, Kaper JB, Vedantam G, Viswanathan V. Enteropathogenic Escherichia coli EspH-Mediated Rho GTPase Inhibition Results in Desmosomal Perturbations. Cell Mol Gastroenterol Hepatol 2018; 6:163-180. [PMID: 30003123 PMCID: PMC6039986 DOI: 10.1016/j.jcmgh.2018.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 04/20/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS The diarrheagenic pathogen, enteropathogenic Escherichia coli (EPEC), uses a type III secretion system to deliver effector molecules into intestinal epithelial cells (IECs). While exploring the basis for the lateral membrane separation of EPEC-infected IECs, we observed infection-induced loss of the desmosomal cadherin desmoglein-2 (DSG2). We sought to identify the molecule(s) involved in, and delineate the mechanisms and consequences of, EPEC-induced DSG2 loss. METHODS DSG2 abundance and localization was monitored via immunoblotting and immunofluorescence, respectively. Junctional perturbations were visualized by electron microscopy, and cell-cell adhesion was assessed using dispase assays. EspH alanine-scan mutants as well as pharmacologic agents were used to evaluate impacts on desmosomal alterations. EPEC-mediated DSG2 loss, and its impact on bacterial colonization in vivo, was assessed using a murine model. RESULTS The secreted virulence protein EspH mediates EPEC-induced DSG2 degradation, and contributes to desmosomal perturbation, loss of cell junction integrity, and barrier disruption in infected IECs. EspH sequesters Rho guanine nucleotide exchange factors and inhibits Rho guanosine triphosphatase signaling; EspH mutants impaired for Rho guanine nucleotide exchange factor interaction failed to inhibit RhoA or deplete DSG2. Cytotoxic necrotizing factor 1, which locks Rho guanosine triphosphatase in the active state, jasplakinolide, a molecule that promotes actin polymerization, and the lysosomal inhibitor bafilomycin A, respectively, rescued infected cells from EPEC-induced DSG2 loss. Wild-type EPEC, but not an espH-deficient strain, colonizes mouse intestines robustly, widens paracellular junctions, and induces DSG2 re-localization in vivo. CONCLUSIONS Our studies define the mechanism and consequences of EPEC-induced desmosomal alterations in IECs. These perturbations contribute to the colonization and virulence of EPEC, and likely related pathogens.
Collapse
Key Words
- A/E, attaching and effacing
- BSA, bovine serum albumin
- CM, calcium and magnesium
- DMEM, Dulbecco's modified Eagle medium
- DSC, desmocollin
- DSG, desmoglein
- DSG2
- Desmoglein
- EPEC
- EPEC, enteropathogenic Escherichia coli
- GEF, guanine nucleotide exchange factors
- GTPase, guanosine triphosphatase
- Host–Pathogen Interaction
- IEC, intestinal epithelial cell
- IF, intermediate filament
- PBS, phosphate-buffered saline
- T3SS, type 3 secretion system
- TER, transepithelial electrical resistance
- TJ, tight junction
- WT, wild-type
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Ross Calvin Monasky
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Bryan Angelo P. Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Al B. Agellon
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
| | - Asad Mansoor
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - James B. Kaper
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Gayatri Vedantam
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- Southern Arizona VA Healthcare System, Tucson, Arizona
| | - V.K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- Correspondence Address correspondence to: V. K. Viswanathan, PhD, School of Animal and Comparative Biomedical Sciences, 1006 E. Lowell, Building 106, Room 231, University of Arizona, Tucson, Arizona 85721. fax: (520) 621-6366.
| |
Collapse
|
45
|
Kuo CJ, Wang ST, Chen CS. Detection of Enterohemorrhagic Escherichia Coli Colonization in Murine Host by Non-invasive In Vivo Bioluminescence System. J Vis Exp 2018. [PMID: 29683443 DOI: 10.3791/56169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Enterohemorrhagic E. coli (EHEC) O157:H7, which is a foodborne pathogen that causesdiarrhea, hemorrhagic colitis (HS), and hemolytic uremic syndrome (HUS), colonize to the intestinal tract of humans. To study the detailed mechanism of EHEC colonization in vivo, it is essential to have animal models to monitor and quantify EHEC colonization. We demonstrate here a mouse-EHEC colonization model by transforming the bioluminescent expressing plasmid to EHEC to monitor and quantify EHEC colonization in living hosts. Animals inoculated with bioluminescence-labeled EHEC show intense bioluminescent signals in mice by detection with a non-invasive in vivo imaging system. After 1 and 2 days post infection, bioluminescent signals could still be detected in infected animals, which suggests that EHEC colonize in hosts for at least 2 days. We also demonstrate that these bioluminescent EHEC locate to mouse intestine, specifically in the cecum and colon, from ex vivo images. This mouse-EHEC colonization model may serve as a tool to advance the current knowledge of the EHEC colonization mechanism.
Collapse
Affiliation(s)
- Cheng-Ju Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University
| | - Sin-Tian Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University
| | - Chang-Shi Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University;
| |
Collapse
|
46
|
Wang J, Lu J, Xie X, Xiong J, Huang N, Wei H, Jiang S, Peng J. Blend of organic acids and medium chain fatty acids prevents the inflammatory response and intestinal barrier dysfunction in mice challenged with enterohemorrhagic Escherichia coli O157:H7. Int Immunopharmacol 2018; 58:64-71. [PMID: 29555328 DOI: 10.1016/j.intimp.2018.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/27/2018] [Accepted: 03/14/2018] [Indexed: 02/07/2023]
Abstract
Impaired epithelial barrier function disrupts immune homeostasis and increases inflammation in intestines, leading to many intestinal diseases. The blend of organic acids (OAs) and medium chain fatty acids (MCFAs) has been shown to have synergistic bactericidal effect. In this study, we demonstrated that two blends of OAs and MCFAs (OM1 and OM2) could prevent the inflammatory response and intestinal barrier dysfunction in enterohemorrhagic Escherichia coli (EHEC)-infected mice. Treatments of OM1 and OM2 significantly reduced the body weight loss and production of IL-6 and TNF-α induced by EHEC. Mice treated with OM1 and OM2 showed decrease in serum D-lactic concentration, DAO activity and bacterial transfer to liver and spleen. Furthermore, OM1 and OM2 increased the expression of tight junction proteins occludin and ZO-1, mucus protein MUC-2, and host defense peptides mBD1, mBD2 and mBD3. Finally, OM1 and OM2 increased the population of Lactobacillus spp. and Bifidobacterium spp., but decreased that of E. coli in the cecum. These findings indicate that OM1 and OM2 may be used to develop a prophylactic agent for intestinal inflammation and injury in enteric pathogen infection.
Collapse
Affiliation(s)
- Jun Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - JinXin Lu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - XiaoWei Xie
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jia Xiong
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Ningning Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Siwen Jiang
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| |
Collapse
|
47
|
Das S, Jayaratne R, Barrett KE. The Role of Ion Transporters in the Pathophysiology of Infectious Diarrhea. Cell Mol Gastroenterol Hepatol 2018; 6:33-45. [PMID: 29928670 PMCID: PMC6007821 DOI: 10.1016/j.jcmgh.2018.02.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Every year, enteric infections and associated diarrhea kill millions of people. The situation is compounded by increases in the number of enteric pathogens that are acquiring resistance to antibiotics, as well as (hitherto) a relative paucity of information on host molecular targets that may contribute to diarrhea. Many forms of diarrheal disease depend on the dysregulation of intestinal ion transporters, and an associated imbalance between secretory and absorptive functions of the intestinal epithelium. A number of major transporters have been implicated in the pathogenesis of diarrheal diseases and thus an understanding of their expression, localization, and regulation after infection with various bacteria, viruses, and protozoa likely will prove critical in designing new therapies. This article surveys our understanding of transporters that are modulated by specific pathogens and the mechanism(s) involved, thereby illuminating targets that might be exploited for new therapeutic approaches.
Collapse
Key Words
- ATP, adenosine triphosphate
- ATPase, adenosine triphosphatase
- CDI, Clostridium difficile infection
- CFTR, cystic fibrosis transmembrane conductance regulator
- CLCA1, chloride channel accessory 1
- CT, cholera toxin
- CXCR2, C-X-C motif chemokine receptor 2
- DRA, down-regulated in adenoma
- Diarrhea
- ENaC, epithelial sodium channel
- EPEC, enteropathogenic Escherichia coli
- ETEC, enterotoxigenic Escherichia coli
- Enteric Pathogen
- Epithelium
- EspG, Escherichia coli secreted protein G
- GPR39, G-protein coupled receptor 39
- Ion Transport
- KCC, potassium-chloride cotransporter
- LPA, lysophosphatidic acid
- LT, heat-labile toxin
- NHE, sodium/hydrogen exchanger
- NHERF2, sodium/hydrogen exchanger regulatory factor 2
- NKCC, sodium-potassium-2 chloride cotransporter
- ORT, oral rehydration therapy
- PKC, protein kinase C
- SGLT1, sodium-glucose cotransporter 1
- SLC, solute carrier
- ST, heat-stabile toxin
- TNF, tumor necrosis factor
- Tcd, Clostridium difficile toxin
- ZnR, zinc sensing receptor
- cAMP, adenosine 3′,5′-cyclic monophosphate
Collapse
Affiliation(s)
- Soumita Das
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California
| | - Rashini Jayaratne
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Kim E. Barrett
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California,Correspondence Address correspondence to: Kim E. Barrett, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0063. fax: (858) 246-1788.
| |
Collapse
|
48
|
Mullineaux-Sanders C, Suez J, Elinav E, Frankel G. Sieving through gut models of colonization resistance. Nat Microbiol 2018; 3:132-140. [PMID: 29358683 DOI: 10.1038/s41564-017-0095-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/08/2017] [Indexed: 12/24/2022]
Abstract
The development of innovative high-throughput genomics and metabolomics technologies has considerably expanded our understanding of the commensal microorganisms residing within the human body, collectively termed the microbiota. In recent years, the microbiota has been reported to have important roles in multiple aspects of human health, pathology and host-pathogen interactions. One function of commensals that has attracted particular interest is their role in protection against pathogens and pathobionts, a concept known as colonization resistance. However, pathogens are also able to sense and exploit the microbiota during infection. Therefore, obtaining a holistic understanding of colonization resistance mechanisms is essential for the development of microbiome-based and microbiome-targeting therapies for humans and animals. Achieving this is dependent on utilizing physiologically relevant animal models. In this Perspective, we discuss the colonization resistance functions of the gut microbiota and sieve through the advantages and limitations of murine models commonly used to study such mechanisms within the context of enteric bacterial infection.
Collapse
Affiliation(s)
- Caroline Mullineaux-Sanders
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK.
| |
Collapse
|
49
|
Marlicz W, Yung DE, Skonieczna-Żydecka K, Loniewski I, van Hemert S, Loniewska B, Koulaouzidis A. From clinical uncertainties to precision medicine: the emerging role of the gut barrier and microbiome in small bowel functional diseases. Expert Rev Gastroenterol Hepatol 2017; 11:961-978. [PMID: 28618973 DOI: 10.1080/17474124.2017.1343664] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the last decade, remarkable progress has been made in the understanding of disease pathophysiology. Many new theories expound on the importance of emerging factors such as microbiome influences, genomics/omics, stem cells, innate intestinal immunity or mucosal barrier complexities. This has introduced a further dimension of uncertainty into clinical decision-making, but equally, may shed some light on less well-understood and difficult to manage conditions. Areas covered: Comprehensive review of the literature on gut barrier and microbiome relevant to small bowel pathology. A PubMed/Medline search from 1990 to April 2017 was undertaken and papers from this range were included. Expert commentary: The scenario of clinical uncertainty is well-illustrated by functional gastrointestinal disorders (FGIDs). The movement towards achieving a better understanding of FGIDs is expressed in the Rome IV guidelines. Novel diagnostic and therapeutic protocols focused on the GB and SB microbiome can facilitate diagnosis, management and improve our understanding of the underlying pathological mechanisms in FGIDs.
Collapse
Affiliation(s)
- Wojciech Marlicz
- a Department of Gastroenterology , Pomeranian Medical University , Szczecin , Poland
| | - Diana E Yung
- b Centre for Liver and Digestive Disorders , Royal Infirmary of Edinburgh , Edinburgh , United Kingdom
| | | | - Igor Loniewski
- c Department of Biochemistry and Human Nutrition , Pomeranian Medical University , Szczecin , Poland.,d Sanprobi Sp. z o.o. Sp. K , Szczecin , Poland
| | | | - Beata Loniewska
- f Department of Neonatal Diseases , Pomeranian Medical University , Szczecin , Poland
| | - Anastasios Koulaouzidis
- g Centre for Liver and Digestive Disorders , Royal Infirmary of Edinburgh , Edinburgh , United Kingdom
| |
Collapse
|
50
|
Yokoyama M, Narita T, Sakurai H, Katsumata-Kato O, Sugiya H, Fujita-Yoshigaki J. Maintenance of claudin-3 expression and the barrier functions of intercellular junctions in parotid acinar cells via the inhibition of Src signaling. Arch Oral Biol 2017; 81:141-150. [DOI: 10.1016/j.archoralbio.2017.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 05/08/2017] [Accepted: 05/14/2017] [Indexed: 11/28/2022]
|