1
|
Hong SM, Baek DH. Diagnostic Procedures for Inflammatory Bowel Disease: Laboratory, Endoscopy, Pathology, Imaging, and Beyond. Diagnostics (Basel) 2024; 14:1384. [PMID: 39001273 PMCID: PMC11241288 DOI: 10.3390/diagnostics14131384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Diagnosing inflammatory bowel disease (IBD) can often be challenging, and differentiating between Crohn's disease and ulcerative colitis can be particularly difficult. Diagnostic procedures for IBD include laboratory tests, endoscopy, pathological tests, and imaging tests. Serological and stool tests can be easily performed in an outpatient setting and provide critical diagnostic clues. Although endoscopy is an invasive procedure, it offers essential diagnostic information and allows for tissue biopsy and therapeutic procedures. Video capsule endoscopy and device-assisted enteroscopy are endoscopic procedures used to evaluate the small bowel. In addition to endoscopy, magnetic resonance imaging, computed tomography, and ultrasound (US) are valuable tools for small bowel assessment. Among these, US is noninvasive and easily utilized, making its use highly practical in daily clinical practice. Endoscopic biopsy aids in the diagnosis of IBD and is crucial for assessing the histological activity of the disease, facilitating a thorough evaluation of disease remission, and aiding in the development of treatment strategies. Recent advances in artificial intelligence hold promise for enhancing various aspects of IBD management, including diagnosis, monitoring, and precision medicine. This review compiles current procedures and promising future tools for the diagnosis of IBD, providing comprehensive insights.
Collapse
Affiliation(s)
- Seung Min Hong
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Dong Hoon Baek
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| |
Collapse
|
2
|
Solà-Ginés M, Miró L, Bellver-Sanchis A, Griñán-Ferré C, Pallàs M, Pérez-Bosque A, Moretó M, Pont L, Benavente F, Barbosa J, Rodríguez C, Polo J. Nutritional, molecular, and functional properties of a novel enzymatically hydrolyzed porcine plasma product. PLoS One 2024; 19:e0301504. [PMID: 38728303 PMCID: PMC11086891 DOI: 10.1371/journal.pone.0301504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/18/2024] [Indexed: 05/12/2024] Open
Abstract
In the present study, an enzymatically hydrolyzed porcine plasma (EHPP) was nutritionally and molecularly characterized. EHPP molecular characterization showed, in contrast to spray-dried plasma (SDP), many peptides with relative molecular masses (Mr) below 8,000, constituting 73% of the protein relative abundance. IIAPPER, a well-known bioactive peptide with anti-inflammatory and antioxidant properties, was identified. In vivo functionality of EHPP was tested in C. elegans and two different mouse models of intestinal inflammation. In C. elegans subjected to lipopolysaccharide exposure, EHPP displayed a substantial anti-inflammatory effect, enhancing survival and motility by 40% and 21.5%, respectively. Similarly, in mice challenged with Staphylococcus aureus enterotoxin B or Escherichia coli O42, EHPP and SDP supplementation (8%) increased body weight and average daily gain while reducing the percentage of regulatory Th lymphocytes. Furthermore, both products mitigated the increase of pro-inflammatory cytokines expression associated with these challenged mouse models. In contrast, some significant differences were observed in markers such as Il-6 and Tnf-α, suggesting that the products may present different action mechanisms. In conclusion, EHPP demonstrated similar beneficial health effects to SDP, potentially attributable to the immunomodulatory and antioxidant activity of its characteristic low Mr bioactive peptides.
Collapse
Affiliation(s)
| | - Lluïsa Miró
- APC Europe S.L.U., Granollers, Spain
- Departament de Bioquímica i Fisiologia (Secció de Fisiologia), Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona (INSA·UB), Barcelona, Spain
| | - Aina Bellver-Sanchis
- Departament de Farmacologia, Toxicologia i Química Terapèutica (Secció de Farmacologia) Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Neurociències (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Christian Griñán-Ferré
- Departament de Farmacologia, Toxicologia i Química Terapèutica (Secció de Farmacologia) Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Neurociències (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Mercè Pallàs
- Departament de Farmacologia, Toxicologia i Química Terapèutica (Secció de Farmacologia) Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Neurociències (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Anna Pérez-Bosque
- Departament de Bioquímica i Fisiologia (Secció de Fisiologia), Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona (INSA·UB), Barcelona, Spain
| | - Miquel Moretó
- Departament de Bioquímica i Fisiologia (Secció de Fisiologia), Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona (INSA·UB), Barcelona, Spain
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, Barcelona, Spain
- Serra Húnter Programe, Generalitat de Catalunya, Barcelona, Spain
| | | | - José Barbosa
- Serra Húnter Programe, Generalitat de Catalunya, Barcelona, Spain
| | | | | |
Collapse
|
3
|
Radisavljevic N, Metcalfe-Roach A, Cirstea M, Tabusi MM, Bozorgmehr T, Bar-Yoseph H, Finlay BB. Microbiota-mediated effects of Parkinson's disease medications on Parkinsonian non-motor symptoms in male transgenic mice. mSphere 2024; 9:e0037923. [PMID: 38078745 PMCID: PMC10826342 DOI: 10.1128/msphere.00379-23] [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: 07/07/2023] [Accepted: 10/24/2023] [Indexed: 01/31/2024] Open
Abstract
Parkinson's disease (PD) is characterized by motor symptoms and a loss of dopaminergic neurons, as well as a variety of non-motor symptoms, including constipation, depression, and anxiety. Recently, evidence has also accumulated for a link between gut microbiota and PD. Most PD patients are on dopamine replacement therapy, primarily a combination of L-DOPA and carbidopa; however, the effect of these medications on the microbiota and non-motor symptoms in PD is still unclear. In this study, we explored the effects of chronic oral treatment with L-DOPA plus carbidopa (LDCD) on the gut microbiota and non-motor symptoms in males of a transgenic mouse model of PD (dbl-PAC-Tg(SNCAA53T);Snca-/-). To further test whether the effects of these PD medications were mediated by the gut microbiota, oral antibiotic treatment (Abx; vancomycin and neomycin) was included both with and without concurrent LDCD treatment. Post-treatment, the gastrointestinal, motor, and behavioral phenotypes were profiled, and fecal, ileal, and jejunal samples were analyzed for gut microbiota composition by 16S sequencing. LDCD treatment was found to improve symptoms of constipation and depression in this model, concurrent with increases in Turicibacter abundance in the ileum. Abx treatment worsened the symptoms of constipation, possibly through decreased levels of short-chain fatty acids and disrupted gut barrier function. LDCD + Abx treatment showed an interaction effect on behavioral symptoms that was also associated with ileal Turicibacter levels. This study demonstrates that, in a mouse model, PD medications and antibiotics affect PD-related non-motor symptoms potentially via the gut microbiota.IMPORTANCEThe motor symptoms of Parkinson's disease (PD) are caused by a loss of dopamine-producing neurons and are commonly treated with dopamine replacement therapy (L-DOPA plus carbidopa). PD has also been associated with altered gut microbiota composition. However, the effects of these PD medications on PD-related non-motor symptoms and the gut microbiota have not been well characterized. This study uses a transgenic mouse model of PD to help resolve medication-induced microbiota alterations from those that are potentially disease relevant within a PD context, and explores how long-term treatment may interact with the gut microbiota to impact non-motor symptoms.
Collapse
Affiliation(s)
- Nina Radisavljevic
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Avril Metcalfe-Roach
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - M Mahebali Tabusi
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tahereh Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haggai Bar-Yoseph
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Molecular Detection of Virulence Factors in Salmonella serovars Isolated from Poultry and Human Samples. Vet Med Int 2023; 2023:1875253. [PMID: 36910894 PMCID: PMC9998162 DOI: 10.1155/2023/1875253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 03/06/2023] Open
Abstract
Salmonellosis is a common infectious disease in humans caused by Salmonella spp., which in recent years has shown an increase in its incidence, with products of avian origin being a common source of transmission. To present a successful infective cycle, there are molecular mechanisms such as virulence factors that provide characteristics that facilitate survival, colonization, and damage to the host. According to this, the study aims to characterize the virulence factors of Salmonella spp. strains isolated from broilers (n = 39) and humans (n = 10). The presence of 24 virulence genes was evaluated using end-point PCR. All the strains of Salmonella spp. isolated from broiler chickens revealed presence of 7/24 (29, 16%) virulence genes (lpfA, csgA, sitC, sipB, sopB, sopE, and sivH). Regarding the strains isolated from cases of gastroenteritis in humans, all strains contained (14/24, 58, 33%) virulence genes (lpfA, csgA, pagC, msgA, spiA, sitC, iroN, sipB, orgA, hilA, sopB, sifA, avrA, and sivH). In summary, the presence of virulence genes in different strains of Salmonella isolated from broilers and humans could be described as bacteria with potential pathogenicity due to the type and number of virulence genes detected. These findings are beneficial for the pathogenic monitoring of Salmonella in Colombia.
Collapse
|
5
|
Lara-Ochoa C, Huerta-Saquero A, Medrano-López A, Deng W, Finlay BB, Martínez-Laguna Y, Puente JL. GrlR, a negative regulator in enteropathogenic E. coli, also represses the expression of LEE virulence genes independently of its interaction with its cognate partner GrlA. Front Microbiol 2023; 14:1063368. [PMID: 36876072 PMCID: PMC9979310 DOI: 10.3389/fmicb.2023.1063368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Enteropathogenic Escherichia coli (EPEC), enterohemorrhagic E. coli (EHEC) and Citrobacter rodentium (CR) belong to a group of pathogens that share the ability to form "attaching and effacing" (A/E) lesions on the intestinal epithelia. A pathogenicity island known as the locus of enterocyte effacement (LEE) contains the genes required for A/E lesion formation. The specific regulation of LEE genes relies on three LEE-encoded regulators: Ler activates the expression of the LEE operons by antagonizing the silencing effect mediated by the global regulator H-NS, GrlA activates ler expression and GrlR represses the expression of the LEE by interacting with GrlA. However, despite the existing knowledge of LEE regulation, the interplay between GrlR and GrlA and their independent roles in gene regulation in A/E pathogens are still not fully understood. Methods To further explore the role that GrlR and GrlA in the regulation of the LEE, we used different EPEC regulatory mutants and cat transcriptional fusions, and performed protein secretion and expression assays, western blotting and native polyacrylamide gel electrophoresis. Results and discussion We showed that the transcriptional activity of LEE operons increased under LEE-repressing growth conditions in the absence of GrlR. Interestingly, GrlR overexpression exerted a strong repression effect over LEE genes in wild-type EPEC and, unexpectedly, even in the absence of H-NS, suggesting that GrlR plays an alternative repressor role. Moreover, GrlR repressed the expression of LEE promoters in a non-EPEC background. Experiments with single and double mutants showed that GrlR and H-NS negatively regulate the expression of LEE operons at two cooperative yet independent levels. In addition to the notion that GrlR acts as a repressor by inactivating GrlA through protein-protein interactions, here we showed that a DNA-binding defective GrlA mutant that still interacts with GrlR prevented GrlR-mediated repression, suggesting that GrlA has a dual role as a positive regulator by antagonizing GrlR's alternative repressor role. In line with the importance of the GrlR-GrlA complex in modulating LEE gene expression, we showed that GrlR and GrlA are expressed and interact under both inducing and repressing conditions. Further studies will be required to determine whether the GrlR alternative repressor function depends on its interaction with DNA, RNA, or another protein. These findings provide insight into an alternative regulatory pathway that GrlR employs to function as a negative regulator of LEE genes.
Collapse
Affiliation(s)
- Cristina Lara-Ochoa
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico.,Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico.,Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Ensenada, Mexico
| | - Abraham Medrano-López
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Wanyin Deng
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Ygnacio Martínez-Laguna
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - José L Puente
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
6
|
McConnel CS, Slanzon GS, Parrish LM, Trombetta SC, Shaw LF, Moore DA, Sischo WM. Transcriptional changes detected in fecal RNA from neonatal dairy calves of different breeds following gastrointestinal disease of varying severity. PLoS One 2022; 17:e0278664. [PMID: 36454999 PMCID: PMC9714867 DOI: 10.1371/journal.pone.0278664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Gastrointestinal (GI) disease is a major health concern in preweaned dairy calves. The objective of this fixed cohort study was to use RNA isolated from preweaned Holstein and Jersey heifer calf feces to study the molecular adaptations to variable clinical GI disease. The study was conducted on a commercial calf ranch in the western U.S. Enrolled calves were assessed twice daily for variations in demeanor, milk intake, and hydration. Fecal consistency scores were recorded at enrollment (day 1), and on the day (day 10) that a fecal sample was collected for differential gene expression (DGE). Calves with diarrhea on either day were classified as having either uncomplicated, localized GI disease (scours), or systemic GI disease (systemic enteritis). Eighty-four calves' fecal RNA was evaluated for DGE, of which 33 calves (n = 20 Holstein; n = 13 Jersey) were consistently healthy. The remaining 51 calves (n = 23 Holstein; n = 28 Jersey) experienced varying severity of GI disease during the sampling window. Genes of interest were related to the inflammatory response (i.e., IFNG, NFKB1, NOD2, TLR2, and TLR4) and cell membrane or cytoplasmic transport (i.e., AQP3, FABP2, KRT8 and SLC5A1). Breed-specific findings indicated that AQP3, IFNG, and TLR4 were upregulated in Holsteins with systemic enteritis, whereas KRT8 was downregulated in systemically affected Jerseys. Holsteins did not appear affected by scours aside from a tendency for DGE of toll-like receptors (TLRs) on the day of diarrhea. However, Jersey calves consistently demonstrated a tendency to upregulate IFNG, NFKB1, and TLR4 when affected with either scours or systemic enteritis. These findings were more pronounced in systemically affected Jersey calves and were observed as a delayed response to both scours and systemic enteritis. These findings support previous observations suggesting that Holstein calves may be better equipped than Jersey calves to rapidly fight pathogen invasion.
Collapse
Affiliation(s)
- C. S. McConnel
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| | - G. S. Slanzon
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - L. M. Parrish
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - S. C. Trombetta
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - L. F. Shaw
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - D. A. Moore
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - W. M. Sischo
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| |
Collapse
|
7
|
Jin X, Yuan B, Liu M, Zhu M, Zhang X, Xie G, Wu W, Wang Z, Xu H, Lv Y, Huang Y, Wang W. Dietary Hermetia illucens Larvae Replacement Alleviates Diarrhea and Improves Intestinal Barrier Function in Weaned Piglets Challenged With Enterotoxigenic Escherichia coli K88. Front Vet Sci 2021; 8:746224. [PMID: 34901243 PMCID: PMC8655791 DOI: 10.3389/fvets.2021.746224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/26/2021] [Indexed: 11/19/2022] Open
Abstract
A high-quality protein substitute, Hermetia illucens (black soldier fly) larvae powder, is rich in protein and often used in animal feed. This study aimed to investigate the feasibility and optimal ratio of replacing fish meal with H. illucens larvae in weaned piglets and to demonstrate the effects on piglets' growth performance, intestinal microflora and immune performance. Forty-eight female weaned piglets were randomly classified into three groups. Each group consisted of eight pens (replicates), with two piglets per pen. Three groups containing different proportions of H. illucens larvae (0, 4, and 8%) were referred to as C, HI4, and HI8. We first designed a 28-day feeding experiment to detect growth performance; after that, the piglets were induced with oral gavage of enterotoxigenic Escherichia coli K88 (ETEC K88) and recording diarrhea on day 29 of the experiment. Samples were taken on the 32nd day to detect the effect of H. illucens larvae on the immune performance of the weaned piglets. H. illucens larvae replacement did not cause any obvious change in the growth performance nether in HI4 nor in HI8 of weaned piglets with 28 d feeding stage. H. illucens larvae could improve the intestinal health of weaned piglets by increasing the content of Lactobacillus and reducing the content of Streptococcus. Compared with C+K88 group, the diarrhea rate was attenuated for the H. illucens supplemented group. The integrity of ileum villi in HI4+K88 and HI8+K88 groups was better than that in C+K88 group, and the villi in C+K88 group were severely damaged. The expression of IL-10, Occludin and Claudin-3 in the intestinal mucosa of the HI4+K88 group and HI8+K88 group were significantly increased (P < 0.05), and the expression of TNF-α was significantly decreased (P < 0.05) compared with the C+K88 group. The results of immunoblotting also validated that the same ETEC K88 treatment of weaned piglets enhanced the expression of tight junction protein in the intestinal mucosa of the H. illucens addition group. ETEC-induced diarrhea will be reduced by the diet of weaned piglets containing H. illucens larvae, ameliorating the immune performance of piglets. Our results indicates that the optimal dosage of H. illucens replacement in weaned piglets is 4%.
Collapse
Affiliation(s)
- Xinxin Jin
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Boyu Yuan
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Mingming Liu
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingqiang Zhu
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Xue Zhang
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Gaijie Xie
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wenxiang Wu
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zifan Wang
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Haidong Xu
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yantao Lv
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yanhua Huang
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wei Wang
- College of Animal Science & Technology, Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
8
|
Zhu C, Ye JL, Yang J, Yang KM, Chen Z, Liang R, Wu XJ, Wang L, Jiang ZY. Differential expression of intestinal ion transporters and water channel aquaporins in young piglets challenged with enterotoxigenic Escherichia coli K88. J Anim Sci 2018; 95:5240-5252. [PMID: 29293799 DOI: 10.2527/jas2017.1806] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The study was to determine whether the expression of genes involved in intestinal water and ion transport would be affected by enterotoxigenic (ETEC) K88 both in vitro and in vivo. First, 36 male piglets (4 d old) were randomly allotted to either the control or the ETEC K88 group. Each group had 6 replicates with 3 piglets per replicate. All piglets were fed with the same diets for 17 d. On d 15, piglets in the ETEC K88 group were challenged with ETEC K88 (serotype O149:K91:K88ac) at 1 × 10 cfu per pig, whereas those in the control group received the same volume of sterile PBS. After being challenged with ETEC K88 for 72 h (d 18), 1 piglet from each replicate was selected for slaughter to collect samples from the jejunum, ileum, and colon. The mRNA expression and protein abundance of cystic fibrosis transmembrane conductance regulator (CFTR) in the ileum and colon were increased compared with that in the control group ( < 0.05). Furthermore, the mRNA expression of () in the ileum and colon was increased by ETEC K88 challenge ( < 0.05), whereas in the jejunum, both its mRNA and protein expression were increased by ETEC K88 treatment ( < 0.05). Additionally, an established porcine intestinal epithelial cell line (IPEC-J2) was used to investigate the effect and possible mechanism of ETEC K88 on expression of water channel aquaporins (AQP) and ion transporters. Cells (1.17 × 10 per well) were grown in 6-well plates and treated with ETEC K88 at a multiplicity of infection of 50:1 for 3 h. The mRNA expression of , , and () in IPEC-J2 cells was reduced after ETEC K88 treatment ( < 0.05). Further analyses using western blotting also demonstrated that ETEC K88 decreased the protein expression of AQP3, AQP9, and AQP11 in IPEC-J2 cells ( < 0.05). Moreover, the phosphorylation levels of protein kinase A (PKA) and cyclic adenosine monophosphate (cAMP)-response element binding protein (CREB) were decreased by ETEC K88 challenge ( < 0.05). The results indicate that ETEC K88 challenge induced differential expression of intestinal ion transporters and AQP in young piglets, probably by regulation of the cAMP-PKA signaling pathway. This study might provide new insights about the importance of fluid homeostasis in control of ETEC-induced diarrhea in young piglets.
Collapse
|
9
|
Rosa F, Busato S, Avaroma FC, Linville K, Trevisi E, Osorio JS, Bionaz M. Transcriptional changes detected in fecal RNA of neonatal dairy calves undergoing a mild diarrhea are associated with inflammatory biomarkers. PLoS One 2018; 13:e0191599. [PMID: 29373601 PMCID: PMC5786293 DOI: 10.1371/journal.pone.0191599] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022] Open
Abstract
After birth, a newborn calf has to adapt to an extrauterine life characterized by several physiological changes. In particular, maturation of the gastrointestinal tract in a new environment loaded with potential pathogens, which can predispose neonatal calves to develop diarrhea, and is a major cause of morbidity and mortality during the first 4 wks of life. We aimed to investigate the inflammatory adaptations at a transcriptomic level in the gastrointestinal (GI) tract to a mild diarrhea in neonatal dairy calves using RNA isolated from fresh fecal samples. Eight newborn Jersey male calves were used from birth to 5 wks of age and housed in individual pens. After birth, calves received 1.9 L of colostrum from their respective dams. Calves had ad-libitum access to water and starter grain (22% CP) and were fed twice daily a total of 5.6 L pasteurized whole milk. Starter intake, body weight (BW), fecal score, withers height (WH), and rectal temperature (RT) were recorded throughout the experiment. Blood samples were collected weekly for metabolic and inflammatory profiling from wk 0 to wk 5. Fresh fecal samples were collected weekly and immediately flash frozen until RNA was extracted using a Trizol-based method, and subsequently, an RT-qPCR analysis was performed. Orthogonal contrasts were used to evaluate linear or quadratic effects over time. Starter intake, BW, and WH increased over time. Fecal score was greatest (2.6 ± 0.3) during wk 2. The concentrations of IL-6, ceruloplasmin, and haptoglobin had a positive quadratic effect with maximal concentrations during wk 2, which corresponded to the maximal fecal score observed during the same time. The concentration of serum amyloid A decreased over time. The mRNA expression of the proinflammatory related genes TLR4, TNFA, IL8, and IL1B had a positive quadratic effect of time. A time effect was observed for the cell membrane sodium-dependent glucose transporter SLC5A1, for the major carbohydrate facilitated transporter SLC2A2, and water transport function AQP3, where SLC5A1 and AQP3 had a negative quadratic effect over time. Our data support the use of the fecal RNA as a noninvasive tool to investigate intestinal transcriptomic profiling of dairy calves experiencing diarrhea, which would be advantageous for future research including nutritional effects and health conditions.
Collapse
Affiliation(s)
- Fernanda Rosa
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Fatima C. Avaroma
- Escuela Agrícola Panamericana El Zamorano, El Zamorano, Francisco Morazán, Honduras
| | - Kali Linville
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, South Dakota, United States of America
| | - Erminio Trevisi
- Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Johan S. Osorio
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
10
|
Elmi A, Dorey A, Watson E, Jagatia H, Inglis NF, Gundogdu O, Bajaj-Elliott M, Wren BW, Smith DGE, Dorrell N. The bile salt sodium taurocholate induces Campylobacter jejuni outer membrane vesicle production and increases OMV-associated proteolytic activity. Cell Microbiol 2017; 20. [PMID: 29205766 DOI: 10.1111/cmi.12814] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023]
Abstract
Campylobacter jejuni, the leading cause of bacterial acute gastroenteritis worldwide, secretes an arsenal of virulence-associated proteins within outer membrane vesicles (OMVs). C. jejuni OMVs contain three serine proteases (HtrA, Cj0511, and Cj1365c) that cleave the intestinal epithelial cell (IEC) tight and adherens junction proteins occludin and E-cadherin, promoting enhanced C. jejuni adhesion to and invasion of IECs. C. jejuni OMVs also induce IECs innate immune responses. The bile salt sodium taurocholate (ST) is sensed as a host signal to coordinate the activation of virulence-associated genes in the enteric pathogen Vibrio cholerae. In this study, the effect of ST on C. jejuni OMVs was investigated. Physiological concentrations of ST do not have an inhibitory effect on C. jejuni growth until the early stationary phase. Coculture of C. jejuni with 0.1% or 0.2% (w/v) ST stimulates OMV production, increasing both lipid and protein concentrations. C. jejuni ST-OMVs possess increased proteolytic activity and exhibit a different protein profile compared to OMVs isolated in the absence of ST. ST-OMVs exhibit enhanced cytotoxicity and immunogenicity to T84 IECs and enhanced killing of Galleria mellonella larvae. ST increases the level of mRNA transcripts of the OMVs-associated serine protease genes and the cdtABC operon that encodes the cytolethal distending toxin. Coculture with ST significantly enhances the OMVs-induced cleavage of E-cadherin and occludin. C. jejuni OMVs also cleave the major endoplasmic reticulum chaperone protein BiP/GRP78 and this activity is associated with the Cj1365c protease. These data suggest that C. jejuni responds to the presence of physiological concentrations of the bile salt ST that increases OMV production and the synthesis of virulence-associated factors that are secreted within the OMVs. We propose that these events contribute to pathogenesis.
Collapse
Affiliation(s)
- Abdi Elmi
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Amber Dorey
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Heena Jagatia
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Mona Bajaj-Elliott
- Infection, Immunity, Inflammation and Physiological Medicine, UCL Institute of Child Health, London, UK
| | - Brendan W Wren
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - David G E Smith
- School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Nick Dorrell
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
11
|
Dubreuil JD. Enterotoxigenic Escherichia coli targeting intestinal epithelial tight junctions: An effective way to alter the barrier integrity. Microb Pathog 2017; 113:129-134. [DOI: 10.1016/j.micpath.2017.10.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/16/2017] [Accepted: 10/20/2017] [Indexed: 01/07/2023]
|
12
|
Attaching and effacing (A/E) lesion formation by enteropathogenic E. coli on human intestinal mucosa is dependent on non-LEE effectors. PLoS Pathog 2017; 13:e1006706. [PMID: 29084270 PMCID: PMC5685641 DOI: 10.1371/journal.ppat.1006706] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 11/14/2017] [Accepted: 10/20/2017] [Indexed: 01/31/2023] Open
Abstract
Enteropathogenic E. coli (EPEC) is a human pathogen that causes acute and chronic pediatric diarrhea. The hallmark of EPEC infection is the formation of attaching and effacing (A/E) lesions in the intestinal epithelium. Formation of A/E lesions is mediated by genes located on the pathogenicity island locus of enterocyte effacement (LEE), which encode the adhesin intimin, a type III secretion system (T3SS) and six effectors, including the essential translocated intimin receptor (Tir). Seventeen additional effectors are encoded by genes located outside the LEE, in insertion elements and prophages. Here, using a stepwise approach, we generated an EPEC mutant lacking the entire effector genes (EPEC0) and intermediate mutants. We show that EPEC0 contains a functional T3SS. An EPEC mutant expressing intimin but lacking all the LEE effectors but Tir (EPEC1) was able to trigger robust actin polymerization in HeLa cells and mucin-producing intestinal LS174T cells. However, EPEC1 was unable to form A/E lesions on human intestinal in vitro organ cultures (IVOC). Screening the intermediate mutants for genes involved in A/E lesion formation on IVOC revealed that strains lacking non-LEE effector/s have a marginal ability to form A/E lesions. Furthermore, we found that Efa1/LifA proteins are important for A/E lesion formation efficiency in EPEC strains lacking multiple effectors. Taken together, these results demonstrate the intricate relationships between T3SS effectors and the essential role non-LEE effectors play in A/E lesion formation on mucosal surfaces. Enteropathogenic E. coli (EPEC) causes diarrhea and generates the attaching and effacing (A/E) lesion in human gut epithelium. A/E lesion formation requires the locus of enterocyte effacement (LEE) in the bacterial genome, which encodes a protein injection system delivering the translocated intimin receptor (Tir), which binds to intimin on the bacterial surface. Intimin-Tir interaction is sufficient for bacterial attachment to epithelial cells in vitro but additional effectors may be needed for A/E lesion formation in the human gut. By generating deletion mutants lacking combinations or the whole repertoire of protein effectors encoded by EPEC, we show that intimin-Tir interaction is not sufficient and reveal an additive role of non-LEE effectors for A/E lesion formation in human intestinal tissue.
Collapse
|
13
|
Pathogen-induced secretory diarrhea and its prevention. Eur J Clin Microbiol Infect Dis 2016; 35:1721-1739. [DOI: 10.1007/s10096-016-2726-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/05/2016] [Indexed: 12/19/2022]
|
14
|
Pelagalli A, Squillacioti C, Mirabella N, Meli R. Aquaporins in Health and Disease: An Overview Focusing on the Gut of Different Species. Int J Mol Sci 2016; 17:ijms17081213. [PMID: 27472320 PMCID: PMC5000611 DOI: 10.3390/ijms17081213] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/14/2016] [Accepted: 07/14/2016] [Indexed: 12/11/2022] Open
Abstract
Aquaporins (AQPs) play a pivotal role in gut homeostasis since their distribution and function is modulated both in physiological and in pathophysiological conditions. The transport of water and solutes through gut epithelia is essential for osmoregulation and digestive and absorptive functions. This passage is regulated by different AQP isoforms and characterized by their peculiar distribution in the gastrointestinal tract. To date, AQP localization has been identified in the gut and associated organs of several mammalian species by different techniques (immunohistochemical, western blotting, and RT-PCR). The present review describes the modulation of AQP expression, distribution, and function in gut pathophysiology. At the same time, the comparative description of AQP in animal species sheds light on the full range of AQP functions and the screening of their activity as transport modulators, diagnostic biomarkers, and drug targets. Moreover, the phenotype of knockout mice for several AQPs and their compensatory role and the use of specific AQP inhibitors have been also reviewed. The reported data could be useful to design future research in both basic and clinical fields.
Collapse
Affiliation(s)
- Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
- Institute of Biostructures and Bioimages, National Research Council, Via De Amicis 95, 80131 Naples, Italy.
| | - Caterina Squillacioti
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Via Veterinaria 1, 80137 Naples, Italy.
| | - Nicola Mirabella
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Via Veterinaria 1, 80137 Naples, Italy.
| | - Rosaria Meli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
15
|
Singh AP, Aijaz S. Enteropathogenic E. coli: breaking the intestinal tight junction barrier. F1000Res 2015; 4:231. [DOI: 10.12688/f1000research.6778.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2015] [Indexed: 11/20/2022] Open
Abstract
Enteropathogenic E. coli (EPEC) causes acute intestinal infections in infants in the developing world. Infection typically spreads through contaminated food and water and leads to severe, watery diarrhea. EPEC attaches to the intestinal epithelial cells and directly injects virulence factors which modulate multiple signaling pathways leading to host cell dysfunction. However, the molecular mechanisms that regulate the onset of diarrhea are poorly defined. A major target of EPEC is the host cell tight junction complex which acts as a barrier and regulates the passage of water and solutes through the paracellular space. In this review, we focus on the EPEC effectors that target the epithelial barrier, alter its functions and contribute to leakage through the tight junctions.
Collapse
|
16
|
Abstract
Enteropathogenic
E. coli (EPEC) causes acute intestinal infections in infants in the developing world. Infection typically spreads through contaminated food and water and leads to severe, watery diarrhea. EPEC attaches to the intestinal epithelial cells and directly injects virulence factors which modulate multiple signaling pathways leading to host cell dysfunction. However, the molecular mechanisms that regulate the onset of diarrhea are poorly defined. A major target of EPEC is the host cell tight junction complex which acts as a barrier and regulates the passage of water and solutes through the paracellular space. In this review, we focus on the EPEC effectors that target the epithelial barrier, alter its functions and contribute to leakage through the tight junctions.
Collapse
Affiliation(s)
- Anand Prakash Singh
- Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Saima Aijaz
- Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
17
|
Sexual maturation and changes in water and salt transport components in the kidney and intestine of three-spined stickleback (Gasterosteus aculeatus L.). Comp Biochem Physiol A Mol Integr Physiol 2015; 188:107-19. [PMID: 26135640 DOI: 10.1016/j.cbpa.2015.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
Abstract
Mature three-spined stickleback males use spiggin threads secreted from their kidney to glue together nest material. This requires strongly hypertrophied renal proximal tubular cells, which compromises renal osmoregulatory function during the breeding period. Experimental evidence suggests that the intestine takes over hypotonic fluid secretion at that stage but the mechanism is unexplored. To unravel the molecular mechanism we analyzed and compared transcript levels of several membrane proteins involved in water and salt transport in intestinal and renal tissues, in non-mature males (NM), mature males (MM), and mature females (MF). Aquaporin paralogs aqp1a, -3a, -8aa, -8ab, -10a, and -10b, two Na(+),K(+)-ATPase alpha-1 subunit isoforms (nka547, nka976), Na(+),K(+),2Cl(-)-, and Na(+),Cl(-)-cotransporters (nkcc1a, nkcc2, ncc), the cystic fibrosis transmembrane conductance regulator (cftr) and two claudin isoforms (cldn2, cldn15a) were expressed in the intestine and kidney in all groups. There were no differences in aqp and cldn expression between intestines of NM and MM; nkcc2 was lower and nka levels tended to be higher in intestines of MM than in NM. In the kidney, aqp1 and aqp8ab levels were lower in MM than in NM, whereas aqp3a, nkcc1a, cldn15a, and spiggin were markedly elevated. This was accompanied by marked hypertrophy of kidney tubules in MM. The data support an altered kidney function in terms of water handling in mature males, whereas there was no support for modified trans-epithelial water permeability or salt-secretory activity in the intestine of mature males. Salt-absorptive activity in the intestine may, however, be down-regulated during male maturation.
Collapse
|
18
|
Cruz-Córdova A, Espinosa-Mazariego K, Ochoa SA, Saldaña Z, Rodea GE, Cázares-Domínguez V, Rodríguez-Ramírez V, Eslava-Campos CA, Navarro-Ocaña A, Arrellano-Galindo J, Hernández-Castro R, Gómez-Duarte OG, Qadri F, Xicohtencatl-Cortes J. CS21 positive multidrug-resistant ETEC clinical isolates from children with diarrhea are associated with self-aggregation, and adherence. Front Microbiol 2014; 5:709. [PMID: 25646093 PMCID: PMC4297921 DOI: 10.3389/fmicb.2014.00709] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/28/2014] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) colonize the human intestinal mucosa using pili and non-pili colonization factors (CFs). CS21 (also designated Longus) is one of the most prevalent CFs encoded by a 14 kb lng DNA cluster located in a virulence plasmid of ETEC; yet limited information is available on the prevalence of CS21 positive ETEC isolates in different countries. The aim of this study was to evaluate the prevalence of CS21 among ETEC clinical isolates from Mexican and Bangladeshi children under 5 years old with diarrhea and to determine the phenotypic and genotypic features of these isolates. METHODS ETEC clinical isolates positive to lngA gene were characterized by genotype, multidrug-resistance, self-aggregation, biofilm formation, and adherence to HT-29 cell line. RESULTS A collection of 303 E. coli clinical isolates were analyzed, the 81.51% (247/303) were identified as ETEC, 30.76% (76/247) were st (+)/lt (+), and 25.10% (62/247) were positive for the lngA gene. Among the lngA (+) ETECs identified, 50% of isolates (31/62) were positive for LngA protein. The most frequent serotype was O128ac:H12 found in 19.35% (12/62) of lngA (+) ETEC studied. Multidrug-resistance (MDR) lngA (+) ETEC isolates was identified in 65% (39/60), self-aggregation in 48.38% (30/62), and biofilm formation in 83.87% (52/62). ETEC lngA (+) isolates were able to adhere to HT-29 cells at different levels. Two lngA isogenic mutants were constructed in the ETEC E9034A and ETEC73332 clinical isolate, showing a 77% and 98% reduction in adherence, respectively with respect to the wild type. CONCLUSION ETEC isolates that have the lngA gene showed features associated with self-aggregation, and adherence to HT-29 cells, important characteristics in the human gut colonization process and pathogenesis.
Collapse
Affiliation(s)
- Ariadnna Cruz-Córdova
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Karina Espinosa-Mazariego
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Sara A Ochoa
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Zeus Saldaña
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico ; Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Gerardo E Rodea
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico ; Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Vicenta Cázares-Domínguez
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Viridiana Rodríguez-Ramírez
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Carlos A Eslava-Campos
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Patogenicidad Bacteriana, Hospital Infantil de México Federico Gómez Mexico City, Mexico ; Departamento de Salud Pública, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Armando Navarro-Ocaña
- Departamento de Salud Pública, Facultad de Medicina, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - José Arrellano-Galindo
- Laboratorio de Infectología, Departamento de Infectología, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| | - Rigoberto Hernández-Castro
- Departamento de Ecología de Agentes Patógenos, Hospital General "Dr. Manuel Gea González," Mexico City, Mexico
| | - Oscar G Gómez-Duarte
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University School of Medicine Nashville, TN, USA
| | - Firdausi Qadri
- Centre for Vaccine Sciences, International Centre for Diarrhoeal Disease Research Dhaka, Bangladesh
| | - Juan Xicohtencatl-Cortes
- Unidad de Hemato-Onocología e Investigación, Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez Mexico City, Mexico
| |
Collapse
|
19
|
Mouse intestinal innate immune responses altered by enterotoxigenic Escherichia coli (ETEC) infection. Microbes Infect 2014; 16:954-61. [DOI: 10.1016/j.micinf.2014.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/04/2014] [Accepted: 09/18/2014] [Indexed: 12/12/2022]
|
20
|
Chen S, Liu S, Zhang F, Ren W, Li N, Yin J, Duan J, Peng Y, Liu G, Yin Y, Wu G. Effects of dietary l-glutamine supplementation on specific and general defense responses in mice immunized with inactivated Pasteurella multocida vaccine. Amino Acids 2014; 46:2365-75. [DOI: 10.1007/s00726-014-1789-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/13/2014] [Indexed: 10/25/2022]
|
21
|
Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 2014; 77:380-439. [PMID: 24006470 DOI: 10.1128/mmbr.00064-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hosts are protected from attack by potentially harmful enteric microorganisms, viruses, and parasites by the polarized fully differentiated epithelial cells that make up the epithelium, providing a physical and functional barrier. Enterovirulent bacteria interact with the epithelial polarized cells lining the intestinal barrier, and some invade the cells. A better understanding of the cross talk between enterovirulent bacteria and the polarized intestinal cells has resulted in the identification of essential enterovirulent bacterial structures and virulence gene products playing pivotal roles in pathogenesis. Cultured animal cell lines and cultured human nonintestinal, undifferentiated epithelial cells have been extensively used for understanding the mechanisms by which some human enterovirulent bacteria induce intestinal disorders. Human colon carcinoma cell lines which are able to express in culture the functional and structural characteristics of mature enterocytes and goblet cells have been established, mimicking structurally and functionally an intestinal epithelial barrier. Moreover, Caco-2-derived M-like cells have been established, mimicking the bacterial capture property of M cells of Peyer's patches. This review intends to analyze the cellular and molecular mechanisms of pathogenesis of human enterovirulent bacteria observed in infected cultured human colon carcinoma enterocyte-like HT-29 subpopulations, enterocyte-like Caco-2 and clone cells, the colonic T84 cell line, HT-29 mucus-secreting cell subpopulations, and Caco-2-derived M-like cells, including cell association, cell entry, intracellular lifestyle, structural lesions at the brush border, functional lesions in enterocytes and goblet cells, functional and structural lesions at the junctional domain, and host cellular defense responses.
Collapse
|
22
|
Awad WA, Hess C, Khayal B, Aschenbach JR, Hess M. In vitro exposure to Escherichia coli decreases ion conductance in the jejunal epithelium of broiler chickens. PLoS One 2014; 9:e92156. [PMID: 24637645 PMCID: PMC3956886 DOI: 10.1371/journal.pone.0092156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 02/17/2014] [Indexed: 02/04/2023] Open
Abstract
Escherichia coli (E. coli) infections are very widespread in poultry. However, little is known about the interaction between the intestinal epithelium and E. coli in chickens. Therefore, the effects of avian non-pathogenic and avian pathogenic Escherichia coli (APEC) on the intestinal function of broiler chickens were investigated by measuring the electrogenic ion transport across the isolated jejunal mucosa. In addition, the intestinal epithelial responses to cholera toxin, histamine and carbamoylcholine (carbachol) were evaluated following an E. coli exposure. Jejunal tissues from 5-week-old broilers were exposed to 6×10(8) CFU/mL of either avian non-pathogenic E. coli IMT11322 (Ont:H16) or avian pathogenic E. coli IMT4529 (O24:H4) in Ussing chambers and electrophysiological variables were monitored for 1 h. After incubation with E. coli for 1 h, either cholera toxin (1 mg/L), histamine (100 μM) or carbachol (100 μM) were added to the incubation medium. Both strains of avian E. coli (non-pathogenic and pathogenic) reduced epithelial ion conductance (Gt) and short-circuit current (Isc). The decrease in ion conductance after exposure to avian pathogenic E. coli was, at least, partly reversed by the histamine or carbachol treatment. Serosal histamine application produced no significant changes in the Isc in any tissues. Only the uninfected control tissues responded significantly to carbachol with an increase of Isc, while the response to carbachol was blunted to non-significant values in infected tissues. Together, these data may explain why chickens rarely respond to intestinal infections with overt secretory diarrhea. Instead, the immediate response to intestinal E. coli infections appears to be a tightening of the epithelial barrier.
Collapse
Affiliation(s)
- Wageha A. Awad
- Department for Farm Animals and Veterinary Public Health, Clinic for Poultry and Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Claudia Hess
- Department for Farm Animals and Veterinary Public Health, Clinic for Poultry and Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Basel Khayal
- Department for Farm Animals and Veterinary Public Health, Clinic for Poultry and Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Faculty of Veterinary Medicine, Free University of Berlin, Berlin, Germany
| | - Michael Hess
- Department for Farm Animals and Veterinary Public Health, Clinic for Poultry and Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
23
|
Real-time sensing of enteropathogenic E. coli-induced effects on epithelial host cell height, cell-substrate interactions, and endocytic processes by infrared surface plasmon spectroscopy. PLoS One 2013; 8:e78431. [PMID: 24194932 PMCID: PMC3806826 DOI: 10.1371/journal.pone.0078431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/19/2013] [Indexed: 12/12/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is an important, generally non-invasive, bacterial pathogen that causes diarrhea in humans. The microbe infects mainly the enterocytes of the small intestine. Here we have applied our newly developed infrared surface plasmon resonance (IR-SPR) spectroscopy approach to study how EPEC infection affects epithelial host cells. The IR-SPR experiments showed that EPEC infection results in a robust reduction in the refractive index of the infected cells. Assisted by confocal and total internal reflection microscopy, we discovered that the microbe dilates the intercellular gaps and induces the appearance of fluid-phase-filled pinocytic vesicles in the lower basolateral regions of the host epithelial cells. Partial cell detachment from the underlying substratum was also observed. Finally, the waveguide mode observed by our IR-SPR analyses showed that EPEC infection decreases the host cell's height to some extent. Together, these observations reveal novel impacts of the pathogen on the host cell architecture and endocytic functions. We suggest that these changes may induce the infiltration of a watery environment into the host cell, and potentially lead to failure of the epithelium barrier functions. Our findings also indicate the great potential of the label-free IR-SPR approach to study the dynamics of host-pathogen interactions with high spatiotemporal sensitivity.
Collapse
|
24
|
Identification of Coli Surface Antigen 23, a novel adhesin of enterotoxigenic Escherichia coli. Infect Immun 2012; 80:2791-801. [PMID: 22645287 DOI: 10.1128/iai.00263-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of diarrhea, mainly in developing countries. Although there are 25 different ETEC adhesins described in strains affecting humans, between 15% and 50% of the clinical isolates from different geographical regions are negative for these adhesins, suggesting that additional unidentified adhesion determinants might be present. Here, we report the discovery of Coli Surface Antigen 23 (CS23), a novel adhesin expressed by an ETEC serogroup O4 strain (ETEC 1766a), which was negative for the previously known ETEC adhesins, albeit it has the ability to adhere to Caco-2 cells. CS23 is encoded by an 8.8-kb locus which contains 9 open reading frames (ORFs), 7 of them sharing significant identity with genes required for assembly of K88-related fimbriae. This gene locus, named aal (adhesion-associated locus), is required for the adhesion ability of ETEC 1766a and was able to confer this adhesive phenotype to a nonadherent E. coli HB101 strain. The CS23 major structural subunit, AalE, shares limited identity with known pilin proteins, and it is more closely related to the CS13 pilin protein CshE, carried by human ETEC strains. Our data indicate that CS23 is a new member of the diverse adhesin repertoire used by ETEC strains.
Collapse
|
25
|
Aroeti B, Friedman G, Zlotkin-Rivkin E, Donnenberg MS. Retraction of enteropathogenic E. coli type IV pili promotes efficient host cell colonization, effector translocation and tight junction disruption. Gut Microbes 2012; 3:267-71. [PMID: 22572833 PMCID: PMC3427219 DOI: 10.4161/gmic.19814] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type IV pili (Tfp) play a primary role in mediating the adherence of pathogenic bacteria to their hosts. The pilus filament can retract with an immense force. However, the role of this activity in microbial pathogenesis has not been rigorously explored. Experiments performed on volunteers suggested that the retraction capacity of enteropathogenic Escherichia coli (EPEC) Tfp is required for full virulence. Here we review our recent study(1) in which we showed that the retraction capacity of the EPEC Tfp facilitates tight-junction disruption and actin-rich pedestal formation by promoting efficient bacterial protein effector translocation into epithelial host cells. We also present new data using live imaging confocal microscopy suggesting that EPEC adheres to monolayers in microcolonies and that Tfp retraction facilitates significant changes in the microcolony shape, which may be critical for efficient effector delivery. Our studies hence suggest novel insights into the role of pili retraction in EPEC pathogenesis.
Collapse
Affiliation(s)
- Benjamin Aroeti
- Department of Cell and Developmental Biology; Institute of Life Sciences; Hebrew University of Jerusalem; Jerusalem, Israel,Correspondence to: Benjamin Aroeti,
| | - Gil Friedman
- Department of Cell and Developmental Biology; Institute of Life Sciences; Hebrew University of Jerusalem; Jerusalem, Israel
| | - Efrat Zlotkin-Rivkin
- Department of Cell and Developmental Biology; Institute of Life Sciences; Hebrew University of Jerusalem; Jerusalem, Israel
| | - Michael S. Donnenberg
- Division of Infectious Diseases; University of Maryland School of Medicine; Baltimore, MD USA
| |
Collapse
|
26
|
Humen MA, Pérez PF, Liévin-Le Moal V. Lipid raft-dependent adhesion of Giardia intestinalis trophozoites to a cultured human enterocyte-like Caco-2/TC7 cell monolayer leads to cytoskeleton-dependent functional injuries. Cell Microbiol 2011; 13:1683-702. [PMID: 21790940 DOI: 10.1111/j.1462-5822.2011.01647.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Gardia intestinalis, the aetiological agent of giardiasis, one of the most common intestinal diseases in both developing and developed countries, induces a loss of epithelial barrier function and functional injuries of the enterocyte by mechanisms that remain unknown. Three possible mechanisms have been proposed: (i) Giardia may directly alter the epithelial barrier after a close interaction between the trophozoite and polarized intestinal cells, (ii) intestinal functions may be altered by factors secreted by Giardia including an 'enterotoxin', proteinases and lectins, and (iii) based on mouse studies, a mechanism involving the intervention of activated T lymphocytes. We used fully differentiated cultured human intestinal Caco-2/TC7 cells forming a monolayer and expressing several polarized functions of enterocytes of small intestine to investigate the mechanisms by which G. intestinalis induces structural and functional alterations in the host intestinal epithelium. We first report that adhesion of G. intestinalis at the brush border of enterocyte-like cells involves the lipid raft membrane microdomains of the trophozoite. We report an adhesion-dependent disorganization of the apical F-actin cytoskeleton that, in turn, results in a dramatic loss of distribution of functional brush border-associated proteins, including sucrase-isomaltase (SI), dipeptidylpeptidase IV (DPP IV) and fructose transporter, GLUT5, and a decrease in sucrose enzyme activity in G. intestinalis-infected enterocyte-like cells. We observed that the G. intestinalis trophozoite promotes an adhesion-dependent decrease in transepithelial electrical resistance (TER) accompanied by a rearrangement of functional tight junction (TJ)-associated occludin, and delocalization of claudin-1. Finally, we found that whereas the occludin rearrangement induced by G. intestinalis was related to apical F-actin disorganization, the delocalization of claudin-1 was not.
Collapse
|
27
|
Zahavi EE, Lieberman JA, Donnenberg MS, Nitzan M, Baruch K, Rosenshine I, Turner JR, Melamed-Book N, Feinstein N, Zlotkin-Rivkin E, Aroeti B. Bundle-forming pilus retraction enhances enteropathogenic Escherichia coli infectivity. Mol Biol Cell 2011; 22:2436-47. [PMID: 21613538 PMCID: PMC3135470 DOI: 10.1091/mbc.e11-01-0001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) and other pathogenic bacteria use dynamic type IV pili to adhere to the host. Here we show that the capacity of the EPEC type IV pili to retract is required for the breakdown of the host epithelial tight-junction barrier, efficient actin-pedestal formation, and translocation of effectors via the type III secretion system. Enteropathogenic Escherichia coli (EPEC) is an important human pathogen that causes acute infantile diarrhea. The type IV bundle-forming pili (BFP) of typical EPEC strains are dynamic fibrillar organelles that can extend out and retract into the bacterium. The bfpF gene encodes for BfpF, a protein that promotes pili retraction. The BFP are involved in bacterial autoaggregation and in mediating the initial adherence of the bacterium with its host cell. Importantly, BFP retraction is implicated in virulence in experimental human infection. How pili retraction contributes to EPEC pathogenesis at the cellular level remains largely obscure, however. In this study, an effort has been made to address this question using engineered EPEC strains with induced BFP retraction capacity. We show that the retraction is important for tight-junction disruption and, to a lesser extent, actin-rich pedestal formation by promoting efficient translocation of bacterial protein effectors into the host cells. A model is proposed whereby BFP retraction permits closer apposition between the bacterial and the host cell surfaces, thus enabling timely and effective introduction of bacterial effectors into the host cell via the type III secretion apparatus. Our studies hence suggest novel insights into the involvement of pili retraction in EPEC pathogenesis.
Collapse
Affiliation(s)
- Eitan E Zahavi
- Department of Cell and Developmental Biology, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Malinowski M, Martus P, Lock JF, Neuhaus P, Stockmann M. Systemic influence of immunosuppressive drugs on small and large bowel transport and barrier function. Transpl Int 2010; 24:184-93. [DOI: 10.1111/j.1432-2277.2010.01167.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
29
|
Collaco A, Marathe J, Kohnke H, Kravstov D, Ameen N. Syntaxin 3 is necessary for cAMP- and cGMP-regulated exocytosis of CFTR: implications for enterotoxigenic diarrhea. Am J Physiol Cell Physiol 2010; 299:C1450-60. [PMID: 20844248 DOI: 10.1152/ajpcell.00029.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Enterotoxins elaborated by Vibrio cholerae and Escherichia coli cannot elicit fluid secretion in the absence of functional cystic fibrosis transmembrane conductance regulator (CFTR) chloride channels. After enterotoxin exposure, CFTR channels are rapidly recruited from endosomes and undergo exocytic insertion into the apical plasma membrane of enterocytes to increase the number of channels on the cell surface by at least fourfold. However, the molecular machinery that orchestrates exocytic insertion of CFTR into the plasma membrane is largely unknown. The present study used immunofluorescence, immunoblotting, surface biotinylation, glutathione S-transferase (GST) pulldown assays, and immunoprecipitation to identify components of the exocytic soluble N-ethylmaleimide (NEM)-sensitive factor attachment receptor (SNARE) vesicle fusion machinery in cyclic nucleotide-activated exocytosis of CFTR in rat jejunum and polarized intestinal Caco-2(BB)e cells. Syntaxin 3, an intestine-specific SNARE, colocalized with CFTR on the apical domain of enterocytes in rat jejunum and polarized Caco-2(BB)e cells. Coimmunoprecipitation and GST binding studies confirmed that syntaxin 3 interacts with CFTR in vivo. Moreover, heat-stable enterotoxin (STa) activated exocytosis of both CFTR and syntaxin 3 to the surface of rat jejunum. Silencing of syntaxin 3 by short hairpin RNA (shRNA) interference abrogated cyclic nucleotide-stimulated exocytosis of CFTR in cells. These observations reveal a new and important role for syntaxin 3 in the pathophysiology of enterotoxin-elicited diarrhea.
Collapse
Affiliation(s)
- Anne Collaco
- Department of Pediatrics and Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
30
|
Abstract
Central to the pathogenesis of many bacterial pathogens is the ability to deliver effector proteins directly into the cells of their eukaryotic host. EspF is one of many effector proteins exclusive to the attaching and effacing pathogen family that includes enteropathogenic (EPEC) and enterohemorrhagic (EHEC) Escherichia coli. Work in recent years has revealed EspF to be one of the most multifunctional effector proteins known, with defined roles in several host cellular processes, including disruption of the epithelial barrier, antiphagocytosis, microvillus effacement, host membrane remodelling, modulation of the cytoskeleton, targeting and disruption of the nucleolus, intermediate filament disruption, cell invasion, mitochondrial dysfunction, apoptosis, and inhibition of several important epithelial transporters. Surprisingly, despite this high number of functions, EspF is a relatively small effector protein, and recent work has begun to decipher the molecular events that underlie its multifunctionality. This review focuses on the activities of EspF within the host cell and discusses recent findings and molecular insights relating to the virulence functions of this fascinating bacterial effector.
Collapse
|
31
|
Reis RSD, Horn F. Enteropathogenic Escherichia coli, Samonella, Shigella and Yersinia: cellular aspects of host-bacteria interactions in enteric diseases. Gut Pathog 2010; 2:8. [PMID: 20649986 PMCID: PMC2921366 DOI: 10.1186/1757-4749-2-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 07/22/2010] [Indexed: 12/18/2022] Open
Abstract
A successful infection of the human intestine by enteropathogenic bacteria depends on the ability of bacteria to attach and colonize the intestinal epithelium and, in some cases, to invade the host cell, survive intracellularly and disseminate from cell to cell. To accomplish these processes bacteria have evolved an arsenal of molecules that are mostly secreted by dedicated type III secretion systems, and that interact with the host, subverting normal cellular functions. Here we overview the most important molecular strategies developed by enteropathogenic Escherichia coli, Salmonella enterica, Shigella flexneri, and Yersinia enterocolitica to cause enteric infections. Despite having evolved different effectors, these four microorganisms share common host cellular targets.
Collapse
Affiliation(s)
- Roberta Souza Dos Reis
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul, P,O, Box 15005, 91501-970, Porto Alegre, Brazil.
| | | |
Collapse
|
32
|
Molecular characterization of GrlA, a specific positive regulator of ler expression in enteropathogenic Escherichia coli. J Bacteriol 2010; 192:4627-42. [PMID: 20622062 DOI: 10.1128/jb.00307-10] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) infections are characterized by the formation of attaching and effacing (A/E) lesions on the surfaces of infected epithelial cells. The genes required for the formation of A/E lesions are located within the locus of enterocyte effacement (LEE). Ler is the key regulatory factor controlling the expression of LEE genes. Expression of the ler gene is positively regulated by GrlA, which is encoded by the LEE. Here, we analyze the mechanism by which GrlA positively regulates ler expression and show that in the absence of H-NS, GrlA is no longer essential for ler activation, further confirming that GrlA acts in part as an H-NS antagonist on the ler promoter. Single-amino-acid mutants were constructed to test the functional significance of the putative helix-turn-helix (HTH) DNA binding motif found in the N-terminal half of GrlA, as well as at the C-terminal domain of the protein. Several mutations within the HTH motif, but not all, completely abolished GrlA activity, as well as specific binding to its target sequence downstream from position -54 in the ler regulatory region. Some of these mutants, albeit inactive, were still able to interact with the negative regulator GrlR, indicating that loss of activity was not a consequence of protein misfolding. Additional residues in the vicinity of the HTH domain, as well as at the end of the protein, were also shown to be important for GrlA activity as a transcriptional regulator, but not for its interaction with GrlR. In summary, GrlA consists of at least two functional domains, one involved in transcriptional activation and DNA binding and the other in heterodimerization with GrlR.
Collapse
|
33
|
Abstract
Current models of Crohn's disease (CD) invoke an initial disturbance of the epithelial interface between the gut mucosa and intestinal microbiota. This "outside-in" paradigm, mirroring the pathophysiology of acute gastroenteritis, suggests that mucosal damage by luminal bacteria is an early, initiating factor in the etiopathogenesis of disease. However, a number of features of CD argue against a primary mucosal process, including phenotypic studies of CD patients that point to a macrophage defect and genetic studies that predict impaired innate immunity to intracellular bacteria. Intracellular pathogens, such as Listeria, Salmonella, and Mycobacteria, invade via the gastrointestinal tract with minimal or no acute mucosal pathology. These organisms then infect and persist in lymphatic tissues before inducing pathology, in the gut or elsewhere, as a secondary process. In a disease resulting from impaired macrophage responses to intracellular pathogens, mucosal damage could instead represent a terminal event in the pathogenesis of disease. Such an "inside-out" model is also compatible with observations on postoperative disease relapses where subepithelial pathology precedes ulceration. This alternative disease paradigm suggests that clinical and experimental research efforts should be directed at deeper processes in the gut wall and attached mesentery to understand how intracellular bacteria could initiate or exacerbate this chronic inflammatory disease.
Collapse
Affiliation(s)
- Marcel A Behr
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
34
|
Bharucha AE, Waldman SA. Taking a lesson from microbial diarrheagenesis in the management of chronic constipation. Gastroenterology 2010; 138:813-7. [PMID: 20114092 PMCID: PMC3141216 DOI: 10.1053/j.gastro.2010.01.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Adil E. Bharucha
- Clinical Enteric Neuroscience Translational and Epidemiological Research Program, Division of Gastroenterology and Hepatology, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Scott A. Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA,To whom correspondences should be addressed: 132 South 10th Street, 1170 Main, Philadelphia, PA, 19107;
| |
Collapse
|