1
|
González-Vergara A, Benavides B, Julio-Pieper M. Mapping and quantifying neuropeptides in the enteric nervous system. J Neurosci Methods 2023; 393:109882. [PMID: 37172914 DOI: 10.1016/j.jneumeth.2023.109882] [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: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Neuropeptides are a highly diverse group of signaling molecules found in the central nervous system (CNS) and peripheral organs, including the enteric nervous system (ENS). Increasing efforts have been focused on dissecting the role of neuropeptides in both neural- and non-neural-related diseases, as well as their potential therapeutic value. In parallel, accurate knowledge on their source of production and pleiotropic functions is still needed to fully understand their implications in biological processes. This review will focus on the analytical challenges involved in studying neuropeptides, particularly in the ENS, a tissue where their abundance is low, together with opportunities for further technical development.
Collapse
Affiliation(s)
- Alex González-Vergara
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Benjamín Benavides
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
2
|
Kaur P, Dudeja PK. Pathophysiology of Enteropathogenic Escherichia coli-induced Diarrhea. NEWBORN (CLARKSVILLE, MD.) 2023; 2:102-113. [PMID: 37388762 PMCID: PMC10308259 DOI: 10.5005/jp-journals-11002-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) are important diarrheal pathogens of infants and young children. Since the availability of molecular diagnosis methods, we now have new insights into the incidence and prevalence of these infections. Recent epidemiological studies indicate that atypical EPEC (aEPEC) are seen more frequently than typical EPEC (tEPEC) worldwide, including in both endemic diarrhea and diarrhea outbreaks. Therefore, it is important to further characterize the pathogenicity of these emerging strains. The virulence mechanisms and pathophysiology of the attaching and effacing lesion (A/E) and the type-three-secretion-system (T3SS) are complex but well-studied. A/E strains use their pool of locus of enterocyte effacement (LEE)-encoded and non-LEE-encoded effector proteins to subvert and modulate cellular and barrier properties of the host. However, the exact mechanisms of diarrhea in EPEC infection are not completely understood. From the clinical perspective, there is a need for fast, easy, and inexpensive diagnostic methods to define optimal treatment and prevention for children in endemic areas. In this article, we present a review of the classification of EPEC, epidemiology, pathogenesis of the disease caused by these bacteria, determinants of virulence, alterations in signaling, determinants of colonization vs. those of disease, and the limited information we have on the pathophysiology of EPEC-induced diarrhea. This article combines peer-reviewed evidence from our own studies and the results of an extensive literature search in the databases PubMed, EMBASE, and Scopus.
Collapse
Affiliation(s)
- Prabhdeep Kaur
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Illinois, United States of America
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
| |
Collapse
|
3
|
Abstract
Enteric bacterial infections contribute substantially to global disease burden and mortality, particularly in the developing world. In vitro 2D monolayer cultures have provided critical insights into the fundamental virulence mechanisms of a multitude of pathogens, including Salmonella enterica serovars Typhimurium and Typhi, Vibrio cholerae, Shigella spp., Escherichia coli and Campylobacter jejuni, which have led to the identification of novel targets for antimicrobial therapy and vaccines. In recent years, the arsenal of experimental systems to study intestinal infections has been expanded by a multitude of more complex models, which have allowed to evaluate the effects of additional physiological and biological parameters on infectivity. Organoids recapitulate the cellular complexity of the human intestinal epithelium while 3D bioengineered scaffolds and microphysiological devices allow to emulate oxygen gradients, flow and peristalsis, as well as the formation and maintenance of stable and physiologically relevant microbial diversity. Additionally, advancements in ex vivo cultures and intravital imaging have opened new possibilities to study the effects of enteric pathogens on fluid secretion, barrier integrity and immune cell surveillance in the intact intestine. This review aims to present a balanced and updated overview of current intestinal in vitro and ex vivo methods for modeling of enteric bacterial infections. We conclude that the different paradigms are complements rather than replacements and their combined use promises to further our understanding of host-microbe interactions and their impacts on intestinal health.
Collapse
Affiliation(s)
- Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- CONTACT Ute Römling Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Volker M. Lauschke Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
4
|
Secombe KR, Van Sebille YZA, Mayo BJ, Coller JK, Gibson RJ, Bowen JM. Diarrhea Induced by Small Molecule Tyrosine Kinase Inhibitors Compared With Chemotherapy: Potential Role of the Microbiome. Integr Cancer Ther 2021; 19:1534735420928493. [PMID: 32493068 PMCID: PMC7273583 DOI: 10.1177/1534735420928493] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Small molecule receptor tyrosine kinase inhibitors (SM-TKIs) are among a group of
targeted cancer therapies, intended to be more specific to cancer cells compared
with treatments, such as chemotherapy, hence reducing adverse events.
Unfortunately, many patients report high levels of diarrhea, the pathogenesis of
which remains under investigation. In this article, we compare the current state
of knowledge of the pathogenesis of chemotherapy-induced diarrhea (CID) in
comparison to SM-TKI–induced diarrhea, and investigate how a similar research
approach in both areas may be beneficial. To this end, we review evidence that
both treatment modalities may interact with the gut microbiome, and as such the
microbiome should be investigated for its ability to reduce the risk of
diarrhea.
Collapse
Affiliation(s)
- Kate R Secombe
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Ysabella Z A Van Sebille
- UniSA Online, Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Bronwen J Mayo
- Division of Health Sciences, University of South Australia, South Australia, Australia
| | - Janet K Coller
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Rachel J Gibson
- School of Allied Health Science and Practice, University of Adelaide, South Australia, Australia
| | - Joanne M Bowen
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Regulatory Influence of Galanin and GALR1/GALR2 Receptors on Inflamed Uterus Contractility in Pigs. Int J Mol Sci 2021; 22:ijms22126415. [PMID: 34203944 PMCID: PMC8232690 DOI: 10.3390/ijms22126415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
Uterine inflammation is a very common and serious pathology in domestic animals, the development and progression of which often result from disturbed myometrial contractility. We investigated the effect of inflammation on the protein expression of galanin (GAL) receptor subtypes (GALR)1 and GALR2 in myometrium and their role in the contractile amplitude and frequency of an inflamed gilt uterus. The gilts of the E. coli and SAL groups received E. coli suspension or saline in their uteri, respectively, and only laparotomy was performed (CON group). Eight days later, the E. coli group developed severe acute endometritis and lowered GALR1 protein expression in the myometrium. Compared to the pretreatment period, GAL (10−7 M) reduced the amplitude and frequency in myometrium and endometrium/myometrium of the CON and SAL groups, the amplitude in both stripes and frequency in endometrium/myometrium of the E. coli group. In this group, myometrial frequency after using GAL increased, and it was higher than in other groups. GALR2 antagonist diminished the decrease in amplitude in myometrium and the frequency in endometrium/myometrium (SAL, E. coli groups) induced by GAL (10−7 M). GALR1/GALR2 antagonist and GAL (10−7 M) reversed the decrease in amplitude and diminished the decrease in frequency in both examined stripes (CON, SAL groups), and diminished the drop in amplitude and abolished the rise in the frequency in the myometrium (E. coli group). In summary, the inflammation reduced GALR1 protein expression in pig myometrium, and GALR1 and GALR2 participated in the contractile regulation of an inflamed uterus.
Collapse
|
6
|
Brzozowska M, Całka J. Review: Occurrence and Distribution of Galanin in the Physiological and Inflammatory States in the Mammalian Gastrointestinal Tract. Front Immunol 2021; 11:602070. [PMID: 33552060 PMCID: PMC7862705 DOI: 10.3389/fimmu.2020.602070] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Galanin (GAL) is a broad-spectrum peptide that was first identified 37 years ago. GAL, which acts through three specific receptor subtypes, is one of the most important molecules on an ever-growing list of neurotransmitters. Recent studies indicate that this peptide is commonly present in the gastrointestinal (GI) tract and GAL distribution can be seen in the enteric nervous system (ENS). The function of the GAL in the gastrointestinal tract is, inter alia, to regulate motility and secretion. It should be noted that the distribution of neuropeptides is largely dependent on the research model, as well as the part of the gastrointestinal tract under study. During the development of digestive disorders, fluctuations in GAL levels were observed. The occurrence of GAL largely depends on the stage of the disease, e.g., in porcine experimental colitis GAL secretion is caused by infection with Brachyspira hyodysenteriae. Many authors have suggested that increased GAL presence is related to the involvement of GAL in organ renewal. Additionally, it is tempting to speculate that GAL may be used in the treatment of gastroenteritis. This review aims to present the function of GAL in the mammalian gastrointestinal tract under physiological conditions. In addition, since GAL is undoubtedly involved in the regulation of inflammatory processes, and the aim of this publication is to provide up-to-date knowledge of the distribution of GAL in experimental models of gastrointestinal inflammation, which may help to accurately determine the role of this peptide in inflammatory diseases and its potential future use in the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Marta Brzozowska
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
7
|
Brunner SM, Reichmann F, Leitner J, Wölfl S, Bereswill S, Farzi A, Schneider AM, Klieser E, Neureiter D, Emberger M, Heimesaat MM, Weghuber D, Lang R, Holzer P, Kofler B. Galanin receptor 3 attenuates inflammation and influences the gut microbiota in an experimental murine colitis model. Sci Rep 2021; 11:564. [PMID: 33436730 PMCID: PMC7803768 DOI: 10.1038/s41598-020-79456-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
The regulatory (neuro)peptide galanin and its three receptors (GAL1-3R) are involved in immunity and inflammation. Galanin alleviated inflammatory bowel disease (IBD) in rats. However, studies on the galanin receptors involved are lacking. We aimed to determine galanin receptor expression in IBD patients and to evaluate if GAL2R and GAL3R contribute to murine colitis. Immunohistochemical analysis revealed that granulocytes in colon specimens of IBD patients (Crohn's disease and ulcerative colitis) expressed GAL2R and GAL3R but not GAL1R. After colitis induction with 2% dextran sulfate sodium (DSS) for 7 days, mice lacking GAL3R (GAL3R-KO) lost more body weight, exhibited more severe colonic inflammation and aggravated histologic damage, with increased infiltration of neutrophils compared to wild-type animals. Loss of GAL3R resulted in higher local and systemic inflammatory cytokine/chemokine levels. Remarkably, colitis-associated changes to the intestinal microbiota, as assessed by quantitative culture-independent techniques, were most pronounced in GAL3R-KO mice, characterized by elevated numbers of enterobacteria and bifidobacteria. In contrast, GAL2R deletion did not influence the course of colitis. In conclusion, granulocyte GAL2R and GAL3R expression is related to IBD activity in humans, and DSS-induced colitis in mice is strongly affected by GAL3R loss. Consequently, GAL3R poses a novel therapeutic target for IBD.
Collapse
MESH Headings
- Animals
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/microbiology
- Colitis, Ulcerative/therapy
- Crohn Disease/genetics
- Crohn Disease/microbiology
- Crohn Disease/therapy
- Gastrointestinal Microbiome
- Gene Expression
- Humans
- Inflammation
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Targeted Therapy
- Rats
- Receptor, Galanin, Type 3/genetics
- Receptor, Galanin, Type 3/metabolism
- Receptor, Galanin, Type 3/physiology
- Mice
Collapse
Affiliation(s)
- Susanne M Brunner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria.
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria
| | - Julia Leitner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Soraya Wölfl
- Laboratory for Pathology Weger, Emberger, Strubergasse 20, 5020, Salzburg, Austria
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Garystr. 5, 14195, Berlin, Germany
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria
| | - Anna-Maria Schneider
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Eckhard Klieser
- Institute of Pathology, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Daniel Neureiter
- Institute of Pathology, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Michael Emberger
- Laboratory for Pathology Weger, Emberger, Strubergasse 20, 5020, Salzburg, Austria
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Garystr. 5, 14195, Berlin, Germany
| | - Daniel Weghuber
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstr. 48, 5020, Salzburg, Austria
| |
Collapse
|
8
|
Serna-Duque JA, Esteban MÁ. Effects of inflammation and/or infection on the neuroendocrine control of fish intestinal motility: A review. FISH & SHELLFISH IMMUNOLOGY 2020; 103:342-356. [PMID: 32454211 DOI: 10.1016/j.fsi.2020.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
Food is the largest expense in fish farms. On the other hand, the fish health and wellbeing are determining factors in aquaculture production where nutrition is a vital process for growing animals. In fact, it is important to remember that digestion and nutrition are crucial for animals' physiology. However, digestion is a very complex process in which food is processed to obtain necessary nutrients and central mechanisms of this process require both endocrine and neuronal regulation. In this context, intestinal motility is essential for the absorption of the nutrients (digestive process determining nutrition). An imbalance in the intestinal motility due to an inadequate diet or an infectious process could result in a lower use of the food and inefficiency in obtaining nutrients from food. Very frequently, farmed fish are infected with different pathogenic microorganism and this situation could alter gastrointestinal physiology and, indirectly reduce fish growth. For these reasons, the present review focuses on analysing how different inflammatory molecules or infections can alter conventional modulators of fish intestinal motility.
Collapse
Affiliation(s)
- Jhon A Serna-Duque
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - M Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain.
| |
Collapse
|
9
|
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
|
10
|
Pearce SC, Coia HG, Karl JP, Pantoja-Feliciano IG, Zachos NC, Racicot K. Intestinal in vitro and ex vivo Models to Study Host-Microbiome Interactions and Acute Stressors. Front Physiol 2018; 9:1584. [PMID: 30483150 PMCID: PMC6240795 DOI: 10.3389/fphys.2018.01584] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome is extremely important for maintaining homeostasis with host intestinal epithelial, neuronal, and immune cells and this host-microbe interaction is critical during times of stress or disease. Environmental, nutritional, and cognitive stress are just a few factors known to influence the gut microbiota and are thought to induce microbial dysbiosis. Research on this bidirectional relationship as it pertains to health and disease is extensive and rapidly expanding in both in vivo and in vitro/ex vivo models. However, far less work has been devoted to studying effects of host-microbe interactions on acute stressors and performance, the underlying mechanisms, and the modulatory effects of different stressors on both the host and the microbiome. Additionally, the use of in vitro/ex vivo models to study the gut microbiome and human performance has not been researched extensively nor reviewed. Therefore, this review aims to examine current evidence concerning the current status of in vitro and ex vivo host models, the impact of acute stressors on gut physiology/microbiota as well as potential impacts on human performance and how we can parlay this information for DoD relevance as well as the broader scientific community. Models reviewed include widely utilized intestinal cell models from human and animal models that have been applied in the past for stress or microbiology research as well as ex vivo organ/tissue culture models and new innovative models including organ-on-a-chip and co-culture models.
Collapse
Affiliation(s)
- Sarah C Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Heidi G Coia
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, Washington, DC, United States.,711th Human Performance Wing, Airforce Research Laboratory, Airman Systems Directorate, Human-Centered ISR Division, Molecular Mechanisms Branch, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - J P Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Ida G Pantoja-Feliciano
- Soldier Protection and Optimization Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kenneth Racicot
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
11
|
Zalecki M, Pidsudko Z, Franke-Radowiecka A, Wojtkiewicz J, Kaleczyc J. Galaninergic intramural nerve and tissue reaction to antral ulcerations. Neurogastroenterol Motil 2018; 30:e13360. [PMID: 29717796 DOI: 10.1111/nmo.13360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/25/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Well-developed galaninergic gastric intramural nerve system is known to regulate multiple stomach functions in physiological and pathological conditions. Stomach ulcer, a disorder commonly occurring in humans and animals, is accompanied by inflammatory reaction. Inflammation can cause intramural neurons to change their neurochemical profile. Galanin and its receptors are involved in inflammation of many organs, however, their direct participation in stomach reaction to ulcer is not known. Therefore, the aim of the study was to investigate adaptive changes in the chemical coding of galaninergic intramural neurons and mRNA expression encoding Gal, GalR1, GalR2, GalR3 receptors in the region of the porcine stomach directly adjacent to the ulcer location. METHODS The experiment was performed on 24 pigs, divided into control and experimental groups. In 12 experimental animals, stomach antrum ulcers were experimentally induced by submucosal injection of acetic acid solution. Stomach wall directly adjacent to the ulcer was examined by: (1) double immunohistochemistry-to verify the changes in the number of galaninergic neurons (submucosal, myenteric) and fibers; (2) real-time PCR to verify changes in mRNA expression encoding galanin, GalR1, GalR2, GalR3 receptors. KEY RESULTS In the experimental animals, the number of Gal-immunoreactive submucosal perikarya was increased, while the number of galaninergic myenteric neurons and fibers (in all the stomach wall layers) remained unchanged. The expression of mRNA encoding all galanin receptors was increased. CONCLUSIONS & INTERFERENCES The results obtained unveiled the participation of galanin and galanin receptors in the stomach tissue response to antral ulcerations.
Collapse
Affiliation(s)
- M Zalecki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Z Pidsudko
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - A Franke-Radowiecka
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - J Wojtkiewicz
- Department of Pathophysiology, Laboratory for Regenerative Medicine, Faculty of Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - J Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
12
|
Chen H, Chen D, Qin W, Liu Y, Che L, Huang Z, Luo Y, Zhang Q, Lin D, Liu Y, Han G, DeSmet S, Michiels J. Wheat bran components modulate intestinal bacteria and gene expression of barrier function relevant proteins in a piglet model. Int J Food Sci Nutr 2016; 68:65-72. [PMID: 27484261 DOI: 10.1080/09637486.2016.1212817] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The objective of this study was to determine the impact of wheat bran and its main polysaccharides on intestinal bacteria and gene expression of intestinal barrier function relevant proteins. Thirty freshly weaned male piglets were assigned randomly to five dietary treatment groups with six piglets per group. Accordingly, five synthetic diets including a basal control diet without fiber components (CON), wheat bran diet (10% wheat bran, WB), arabinoxylan diet (AX), cellulose diet (CEL) and combined diet of arabinoxylan and cellulose (CB) were studied. The piglets were fed ad libitum for 30 d. Lower Escherichia coli (E. coli) populations in WB group and higher probiotic (Lactobacillus and Bifidobacterium) populations in groups fed diets containing arabinoxylan (WB, AX and CB) were observed and compared with CON group. Compared with CON group, the gene expressions of cystic fibrosis transmembrane conductance regulator (CFTR), calcium-activated chloride channel regulator 1 (CLCA1) and voltage-gated chloride channel 2 (CIC2) were suppressed in the WB group. And wheat bran down-regulated gene expression of pro-inflammation (TNF-α, IL-1β, IL-6) and TLRs/MyD88/NF-κB pathway compared with CON group. In conclusion, wheat bran and its main polysaccharides could change intestinal microflora and down-regulate the gene expression of intestinal barrier function relevant proteins in the distal small intestinal mucosa.
Collapse
Affiliation(s)
- Hong Chen
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China.,b Department of Animal Production, Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO) , Ghent University , Melle , Belgium
| | - Daiwen Chen
- c Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Wen Qin
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Yuntao Liu
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Lianqiang Che
- c Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Zhiqing Huang
- c Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Yuheng Luo
- c Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Qing Zhang
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Derong Lin
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Yaowen Liu
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Guoquan Han
- a College of Food Science, Sichuan Agricultural University , Ya'an , Sichuan , PR China
| | - Stefaan DeSmet
- b Department of Animal Production, Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO) , Ghent University , Melle , Belgium
| | - Joris Michiels
- b Department of Animal Production, Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO) , Ghent University , Melle , Belgium.,d Department of Applied Biosciences , Ghent University , Gent , Belgium
| |
Collapse
|
13
|
Zalecki M, Sienkiewicz W, Franke-Radowiecka A, Klimczuk M, Kaleczyc J. The Influence of Gastric Antral Ulcerations on the Expression of Galanin and GalR1, GalR2, GalR3 Receptors in the Pylorus with Regard to Gastric Intrinsic Innervation of the Pyloric Sphincter. PLoS One 2016; 11:e0155658. [PMID: 27175780 PMCID: PMC4866767 DOI: 10.1371/journal.pone.0155658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/02/2016] [Indexed: 01/29/2023] Open
Abstract
Gastric antrum ulcerations are common disorders occurring in humans and animals. Such localization of ulcers disturbs the gastric emptying process, which is precisely controlled by the pylorus. Galanin (Gal) and its receptors are commonly accepted to participate in the regulation of inflammatory processes and neuronal plasticity. Their role in the regulation of gastrointestinal motility is also widely described. However, there is lack of data considering antral ulcerations in relation to changes in the expression of Gal and GalR1, GalR2, GalR3 receptors in the pyloric wall tissue and galaninergic intramural innervation of the pylorus. Two groups of pigs were used in the study: healthy gilts and gilts with experimentally induced antral ulcers. By double immunocytochemistry percentages of myenteric and submucosal neurons expressing Gal-immunoreactivity were determined in the pyloric wall tissue and in the population of gastric descending neurons supplying the pyloric sphincter (labelled by retrograde Fast Blue neuronal tracer). The percentage of Gal-immunoreactive neurons increased only in the myenteric plexus of the pyloric wall (from 16.14±2.06% in control to 25.5±2.07% in experimental animals), while no significant differences in other neuronal populations were observed between animals of both groups. Real-Time PCR revealed the increased expression of mRNA encoding Gal and GalR1 receptor in the pyloric wall tissue of the experimental animals, while the expression(s) of GalR2 and GalR3 were not significantly changed. The results obtained suggest the involvement of Gal, GalR1 and galaninergic pyloric myenteric neurons in the response of pyloric wall structures to antral ulcerations.
Collapse
Affiliation(s)
- Michal Zalecki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- * E-mail:
| | - Waldemar Sienkiewicz
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Amelia Franke-Radowiecka
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Magdalena Klimczuk
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
14
|
Interactions Between Bacteria and the Gut Mucosa: Do Enteric Neurotransmitters Acting on the Mucosal Epithelium Influence Intestinal Colonization or Infection? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:121-41. [DOI: 10.1007/978-3-319-20215-0_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Lang R, Gundlach AL, Holmes FE, Hobson SA, Wynick D, Hökfelt T, Kofler B. Physiology, signaling, and pharmacology of galanin peptides and receptors: three decades of emerging diversity. Pharmacol Rev 2015; 67:118-75. [PMID: 25428932 DOI: 10.1124/pr.112.006536] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Galanin was first identified 30 years ago as a "classic neuropeptide," with actions primarily as a modulator of neurotransmission in the brain and peripheral nervous system. Other structurally-related peptides-galanin-like peptide and alarin-with diverse biologic actions in brain and other tissues have since been identified, although, unlike galanin, their cognate receptors are currently unknown. Over the last two decades, in addition to many neuronal actions, a number of nonneuronal actions of galanin and other galanin family peptides have been described. These include actions associated with neural stem cells, nonneuronal cells in the brain such as glia, endocrine functions, effects on metabolism, energy homeostasis, and paracrine effects in bone. Substantial new data also indicate an emerging role for galanin in innate immunity, inflammation, and cancer. Galanin has been shown to regulate its numerous physiologic and pathophysiological processes through interactions with three G protein-coupled receptors, GAL1, GAL2, and GAL3, and signaling via multiple transduction pathways, including inhibition of cAMP/PKA (GAL1, GAL3) and stimulation of phospholipase C (GAL2). In this review, we emphasize the importance of novel galanin receptor-specific agonists and antagonists. Also, other approaches, including new transgenic mouse lines (such as a recently characterized GAL3 knockout mouse) represent, in combination with viral-based techniques, critical tools required to better evaluate galanin system physiology. These in turn will help identify potential targets of the galanin/galanin-receptor systems in a diverse range of human diseases, including pain, mood disorders, epilepsy, neurodegenerative conditions, diabetes, and cancer.
Collapse
Affiliation(s)
- Roland Lang
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Andrew L Gundlach
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Fiona E Holmes
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Sally A Hobson
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - David Wynick
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Tomas Hökfelt
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Barbara Kofler
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| |
Collapse
|
16
|
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: 4.7] [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
|
17
|
Mahmood F, Hakimiyan A, Jayaraman V, Wood S, Sivaramakrishnan G, Rehman T, Reuhs BL, Chubinskaya S, Shafikhani SH. A novel human antimicrobial factor targets Pseudomonas aeruginosa through its type III secretion system. J Med Microbiol 2013; 62:531-539. [PMID: 23288430 DOI: 10.1099/jmm.0.051227-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic bacterial pathogen. Despite its metabolic and virulence versatility, it has not been shown to infect articular joints, which are areas that are rarely infected with bacteria in general. We hypothesized that articular joints possess antimicrobial activity that limits bacterial survival in these environments. We report that cartilages secrete a novel antimicrobial factor, henceforth referred to as the cartilage-associated antimicrobial factor (CA-AMF), with potent antimicrobial activity. Importantly, CA-AMF exhibited significantly more antimicrobial activity against P. aeruginosa strains with a functional type III secretion system (T3SS). We propose that CA-AMF represents a new class of human antimicrobial factors in innate immunity, one which has evolved to selectively target pathogenic bacteria among the beneficial and commensal microflora. The T3SS is the first example, to the best of our knowledge, of a pathogen-specific molecular target in this antimicrobial defence system.
Collapse
Affiliation(s)
- Fareeha Mahmood
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Arnavaz Hakimiyan
- Department of Food Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Vijayakumar Jayaraman
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Stephen Wood
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | | | - Tooba Rehman
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bradley L Reuhs
- Department of Food Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Susanna Chubinskaya
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H Shafikhani
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
18
|
Nutrition and pathology of weaner pigs: Nutritional strategies to support barrier function in the gastrointestinal tract. Anim Feed Sci Technol 2012. [DOI: 10.1016/j.anifeedsci.2011.12.022] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
19
|
Abstract
The immune system defends the organism against invading pathogens. In recent decades it became evident that elimination of such pathogens, termination of inflammation, and restoration of host homeostasis all depend on bidirectional crosstalk between the immune system and the neuroendocrine system. This crosstalk is mediated by a complex network of interacting molecules that modulates inflammation and cell growth. Among these mediators are neuropeptides released from neuronal and non-neuronal components of the central and peripheral nervous systems, endocrine tissues, and cells of the immune system. Neuropeptide circuitry controls tissue inflammation and maintenance, and an imbalance of pro- and anti-inflammatory neuropeptides results in loss of host homeostasis and triggers inflammatory diseases. The galanin peptide family is undoubtedly involved in the regulation of inflammatory processes, and the aim of this review is to provide up-to-date knowledge from the literature concerning the regulation of galanin and its receptors in the nervous system and peripheral tissues in experimental models of inflammation. We also highlight the effects of galanin and other members of the galanin peptide family on experimentally induced inflammation and discuss these data in light of an anti-inflammatory role for this family of peptides.
Collapse
Affiliation(s)
- Roland Lang
- Department of Dermatology, Paracelsus Medical University Salzburg, Muellner-Hauptstrasse 48, A-5020 Salzburg, Austria
| | | |
Collapse
|
20
|
Roxas JL, Koutsouris A, Bellmeyer A, Tesfay S, Royan S, Falzari K, Harris A, Cheng H, Rhee KJ, Hecht G. Enterohemorrhagic E. coli alters murine intestinal epithelial tight junction protein expression and barrier function in a Shiga toxin independent manner. J Transl Med 2010; 90:1152-68. [PMID: 20479715 PMCID: PMC2912457 DOI: 10.1038/labinvest.2010.91] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Shiga toxin (Stx) is implicated in the development of hemorrhagic colitis and hemolytic-uremic syndrome, but early symptoms of enterohemorrhagic Escherichia coli (EHEC) infection such as nonbloody diarrhea may be Stx independent. In this study, we defined the effects of EHEC, in the absence of Stx, on the intestinal epithelium using a murine model. EHEC colonization of intestines from two groups of antibiotic-free and streptomycin-treated C57Bl/6J mice were characterized and compared. EHEC colonized the cecum and colon more efficiently than the ileum in both groups; however, greater amounts of tissue-associated EHEC were detected in streptomycin-pretreated mice. Imaging of intestinal tissues of mice infected with bioluminescent EHEC further confirmed tight association of the bacteria with the cecum and colon. Greater numbers of EHEC were also cultured from stool samples obtained from streptomycin-pretreated mice, as compared with those that received no antibiotics. Transmission electron microscopy shows that EHEC infection leads to microvillous effacement of mouse colonocytes. Hematoxylin and eosin staining of the colonic tissues of infected mice revealed a slight increase in the number of lamina propria polymorphonuclear leukocytes. Transmucosal electrical resistance, a measure of epithelial barrier function, was reduced in the colonic tissues of infected animals. Increased mucosal permeability to 4- kDa FITC-dextran was also observed in the colonic tissues of infected mice. Immunofluorescence microscopy showed that EHEC infection resulted in redistribution of the tight junction (TJ) proteins occludin and claudin-3 and increased the expression of claudin-2, whereas ZO-1 localization remained unaltered. Quantitative real-time PCR showed that EHEC altered mRNA transcription of OCLN, CLDN2, and CLDN3. Most notably, claudin-2 expression was significantly increased and correlated with increased intestinal permeability. Our data indicate that C57Bl/6J mice serve as an in vivo model to study the physiological effects of EHEC infection on the intestinal epithelium and suggest that altered transcription of TJ proteins has a role in the increase in intestinal permeability.
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Athanasia Koutsouris
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Amy Bellmeyer
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Samuel Tesfay
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Sandhya Royan
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Kanakeshwari Falzari
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Antoneicka Harris
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Hao Cheng
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Ki-Jong Rhee
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Gail Hecht
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA, Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
21
|
Hempson SJ, Matkowskyj K, Bansal A, Tsao E, Habib I, Benya R, Mackow ER, Shaw RD. Rotavirus infection of murine small intestine causes colonic secretion via age restricted galanin-1 receptor expression. Gastroenterology 2010; 138:2410-7. [PMID: 20197068 DOI: 10.1053/j.gastro.2010.02.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 02/01/2010] [Accepted: 02/16/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND & AIMS Mechanisms for age restriction of rotavirus diarrhea are unclear. Because rotavirus primarily infects small intestine, colonic contribution has not been widely studied. Recent data suggest that colonic secretion postbacterial infection is mediated by galanin-1 receptors (Gal1-R). We evaluated age-dependent expression of Gal1-R in Rhesus rotavirus (RRV)-infected mice and its contribution to fluid secretion. METHODS Twenty-four hours after infection of C57BL/6J mice (wild type or Gal1-R knockout) with RRV or vehicle, closed small intestinal and colon loops were constructed. Net fluid content of the loops was calculated (milligrams/centimeters) at 2 hours post-treatment with galanin, galanin antibody, or lidocaine. Gal1-R expression was quantified by automated chromogen analysis. RESULTS Viral antigen was detected in small intestinal epithelial cells but not in colon. Developmental Gal1-R was widely expressed in the small intestine but minimally in the colon. Postinfection, markedly increased Gal1-R was seen in the colon but not after day 25. Galanin caused a significantly higher increase in the net fluid content of infected colon than small intestine. Treatment with lidocaine reduced net fluid secretion in the small intestine and the colon. Mean diarrheal scores were significantly reduced in Gal1-R knockout mice compared with wild type (1.19 +/- 0.31, n = 22 vs 3.36 +/- 0.50, n = 35, P = .0001). CONCLUSIONS These data show that RRV infection of the small intestine increases colonic secretion through Gal1-R and provide a promising start toward understanding the age restriction of rotavirus diarrhea.
Collapse
Affiliation(s)
- Scott J Hempson
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Kolachala VL, Wang L, Obertone TS, Prasad M, Yan Y, Dalmasso G, Gewirtz AT, Merlin D, Sitaraman SV. Adenosine 2B receptor expression is post-transcriptionally regulated by microRNA. J Biol Chem 2010; 285:18184-90. [PMID: 20388705 DOI: 10.1074/jbc.m109.066555] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have reported that epithelial adenosine 2B receptor (A(2B)AR) mRNA and protein are up-regulated in colitis, which we demonstrated to be regulated by tumor necrosis factor alpha (TNF-alpha). Here, we examined the mechanism that governs A(2B)AR expression during colitis. A 1.4-kb sequence of the A(2B)AR promoter was cloned into the pFRL7 luciferase vector. Anti-microRNA (miRNA) was custom-synthesized based on specific miRNA binding sites. The binding of miRNA to the 3'-untranslated region (UTR) of A(2B)AR mRNA was examined by cloning this 3'-UTR downstream of the luciferase gene in pMIR-REPORT. In T84 cells, TNF-alpha induced a 35-fold increase in A(2B)AR mRNA but did not increase promoter activity in luciferase assays. By nuclear run-on assay, no increase in A(2B)AR mRNA following TNF-alpha treatment was observed. Four putative miRNA target sites (miR27a, miR27b, miR128a, miR128b) in the 3'-UTR of the A(2B)AR mRNA were identified in T84 cells and mouse colon. Pretreatment of cells with TNF-alpha reduced the levels of miR27b and miR128a by 60%. Over expression of pre-miR27b and pre-miR128a reduced A(2B)AR levels by >60%. Blockade of miR27b increased A(2B)AR mRNA levels by 6-fold in vitro. miR27b levels declined significantly in colitis-affected tissue in mice in the presence of increased A(2B)AR mRNA. Collectively, these data demonstrate that TNF-alpha-induced A(2B)AR expression in colonic epithelial cells is post-transcriptionally regulated by miR27b and miR128a and show that miR27b influences A(2B)AR expression in murine colitis.
Collapse
Affiliation(s)
- Vasantha L Kolachala
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nadler C, Baruch K, Kobi S, Mills E, Haviv G, Farago M, Alkalay I, Bartfeld S, Meyer TF, Ben-Neriah Y, Rosenshine I. The type III secretion effector NleE inhibits NF-kappaB activation. PLoS Pathog 2010; 6:e1000743. [PMID: 20126447 PMCID: PMC2813277 DOI: 10.1371/journal.ppat.1000743] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 12/29/2009] [Indexed: 11/18/2022] Open
Abstract
The complex host-pathogen interplay involves the recognition of the pathogen by the host's innate immune system and countermeasures taken by the pathogen. Detection of invading bacteria by the host leads to rapid activation of the transcription factor NF-kappaB, followed by inflammation and eradication of the intruders. In response, some pathogens, including enteropathogenic Escherichia coli (EPEC), acquired means of blocking NF-kappaB activation. We show that inhibition of NF-kappaB activation by EPEC involves the injection of NleE into the host cell. Importantly, we show that NleE inhibits NF-kappaB activation by preventing activation of IKKbeta and consequently the degradation of the NF-kappaB inhibitor, IkappaB. This NleE activity is enhanced by, but is not dependent on, a second injected effector, NleB. In conclusion, this study describes two effectors, NleB and NleE, with no similarity to other known proteins, used by pathogens to manipulate NF-kappaB signaling pathways.
Collapse
Affiliation(s)
- Chen Nadler
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Kobi Baruch
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Simi Kobi
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Erez Mills
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Gili Haviv
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Marganit Farago
- Department of Immunology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Irit Alkalay
- Department of Immunology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Sina Bartfeld
- Department Molecular Biology, MPI for Infection Biology, Berlin, Germany
| | - Thomas F. Meyer
- Department Molecular Biology, MPI for Infection Biology, Berlin, Germany
| | - Yinon Ben-Neriah
- Department of Immunology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Ilan Rosenshine
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
24
|
Heuvelin E, Lebreton C, Bichara M, Cerf-Bensussan N, Heyman M. A Bifidobacterium probiotic strain and its soluble factors alleviate chloride secretion by human intestinal epithelial cells. J Nutr 2010; 140:7-11. [PMID: 19889806 DOI: 10.3945/jn.109.114553] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Previous studies indicate that certain probiotic bacterial strains or their soluble products can alleviate proinflammatory cytokine secretion by intestinal epithelial cells (IEC), but their impact on epithelial chloride (Cl(-)) secretion remains elusive. To further decipher the mechanisms of the cross-talk between bacteria/soluble factors and epithelial cells, we analyzed the capacity of the probiotic strain Bifidobacterium breve C50 (Bb C50), its conditioned medium, and other commensal Gram (+) bacteria to modulate epithelial Cl(-) secretion. The effect of Bb C50 on carbachol- (CCh) or forskolin (Fsk)-induced Cl(-) secretion was measured in an IEC line in Ussing chambers. The mechanisms involved in the regulation of Cl(-) secretion were assessed by measuring intracellular Ca(2+) concentration, phosphatase activity, protein kinase (PK) C and PKA activation, and cystic fibrosis transmembrane conductance regulator (CFTR) expression. CCh- or Fsk-induced Cl(-) secretion [short-circuit current (Isc): 151 +/- 28 and 98 +/- 14 microA/cm(2), respectively] was inhibited dose-dependently by Bb C50 (Isc 33 +/- 12 and 49 +/- 7 microA/cm(2) at multiplicity of infection 100; P < 0.02). Fsk-induced Cl(-) secretion was also inhibited by Lactobacillus rhamnosus 10893. No other inhibitory effect was recorded with the other Gram (+) bacteria tested. The inhibitory effect of Bb C50 on CCh-induced Cl(-) secretion targeted a step downstream of epithelial Ca(2+) mobilization and was associated with decreased PKC activity. Thus, Bb C50 and secreted soluble factors, by inhibiting phosphorylation processes, may promote intestinal homeostasis by controlling Cl(-) secretion.
Collapse
|
25
|
Ashokkumar B, Kumar JS, Hecht GA, Said HM. Enteropathogenic Escherichia coli inhibits intestinal vitamin B1 (thiamin) uptake: studies with human-derived intestinal epithelial Caco-2 cells. Am J Physiol Gastrointest Liver Physiol 2009; 297:G825-33. [PMID: 19628653 PMCID: PMC2763801 DOI: 10.1152/ajpgi.00250.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 07/20/2009] [Indexed: 01/31/2023]
Abstract
Infection with the gram-negative enteropathogenic Escherichia coli (EPEC), a food-borne pathogen, represents a significant risk to human health. Whereas diarrhea is a major consequence of this infection, malnutrition also occurs especially in severe and prolonged cases, which may aggravate the health status of the infected hosts. Here we examined the effect of EPEC infection on the intestinal uptake of the water-soluble vitamin B1 (thiamin) using an established human intestinal epithelial Caco-2 cell model. The results showed that infecting Caco-2 cells with wild-type EPEC (but not with nonpathogenic E. coli, killed EPEC, or filtered supernatant) leads to a significant (P < 0.01) inhibition in thiamin uptake. Kinetic parameters of both the nanomolar (mediated by THTR-2) and the micromolar (mediated by THTR-1) saturable thiamin uptake processes were affected by EPEC infection. Cell surface expression of hTHTR-1 and -2 proteins, (determined by the biotinylation method) showed a significantly (P < 0.01) lower expression in EPEC-treated cells compared with controls. EPEC infection also affected the steady-state mRNA levels as well as promoter activity of the SLC19A2 and SLC19A3 genes. Infecting Caco-2 cells with EPEC mutants that harbor mutations in the escN gene (which encodes a putative ATPase for the EPEC type III secretion system, TTSS) or the espA, espB, or espD genes (which encode structural components of the TTSS) did not affect thiamin uptake. On the other hand, mutations in espF and espH genes (which encode effector proteins) exhibited partial inhibition in thiamin uptake. These results demonstrate for the first time that EPEC infection of human intestinal epithelial cells leads to inhibition in thiamin uptake via effects on physiological and molecular parameters of hTHTR-1 and -2. Furthermore, the inhibition appears to be dependent on a functional TTSS of EPEC.
Collapse
Affiliation(s)
- Balasubramaniem Ashokkumar
- Department of Medical Research, Veterans Administration Medical Center, Long Beach, California 90822, USA
| | | | | | | |
Collapse
|
26
|
Lapointe TK, O'Connor PM, Buret AG. The role of epithelial malfunction in the pathogenesis of enteropathogenic E. coli-induced diarrhea. J Transl Med 2009; 89:964-70. [PMID: 19620958 DOI: 10.1038/labinvest.2009.69] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The homeostatic balance of the gastrointestinal tract relies on a single layer of epithelial cells, which assumes both digestive and protective functions. Enteric pathogens, including enteropathogenic Escherichia coli (EPEC), have evolved numerous mechanisms to disrupt basic intestinal epithelial functions, promoting the development of gastrointestinal disorders. Despite its non-invasive nature, EPEC inflicts severe damage to the intestinal mucosa, including the dysregulation of water and solute transport and the disruption of epithelial barrier structure and function. Despite the high prevalence and morbidity of disease caused by EPEC infections, the etiology of its pathogenesis remains incompletely understood. This review integrates the newest findings on EPEC-epithelial interactions with established mechanisms of disease in an attempt to give a comprehensive understanding of the cellular processes whereby this common pathogen may cause diarrheal illness.
Collapse
Affiliation(s)
- Tamia K Lapointe
- Department of Biological Sciences and Inflammation Research Network, University of Calgary, Calgary, AB, Canada
| | | | | |
Collapse
|
27
|
Spehlmann ME, Dann SM, Hruz P, Hanson E, McCole DF, Eckmann L. CXCR2-dependent mucosal neutrophil influx protects against colitis-associated diarrhea caused by an attaching/effacing lesion-forming bacterial pathogen. THE JOURNAL OF IMMUNOLOGY 2009; 183:3332-43. [PMID: 19675161 DOI: 10.4049/jimmunol.0900600] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) is a major cause of diarrheal disease in young children, yet symptoms and duration are highly variable for unknown reasons. Citrobacter rodentium, a murine model pathogen that shares important functional features with EPEC, colonizes mice in colon and cecum and causes inflammation, but typically little or no diarrhea. We conducted genome-wide microarray studies to define mechanisms of host defense and disease in C. rodentium infection. A significant fraction of the genes most highly induced in the colon by infection encoded CXC chemokines, particularly CXCL1/2/5 and CXCL9/10, which are ligands for the chemokine receptors CXCR2 and CXCR3, respectively. CD11b(+) dendritic cells were the major producers of CXCL1, CXCL5, and CXCL9, while CXCL2 was mainly induced in macrophages. Infection of gene-targeted mice revealed that CXCR3 had a significant but modest role in defense against C. rodentium, whereas CXCR2 had a major and indispensable function. CXCR2 was required for normal mucosal influx of neutrophils, which act as direct antibacterial effectors. Moreover, CXCR2 loss led to severe diarrhea and failure to express critical components of normal ion and fluid transport, including ATPase beta(2)-subunit, CFTR, and DRA. The antidiarrheal functions were unique to CXCR2, since other immune defects leading to increased bacterial load and inflammation did not cause diarrhea. Thus, CXCR2-dependent processes, particularly mucosal neutrophil influx, not only contribute to host defense against C. rodentium, but provide protection against infection-associated diarrhea.
Collapse
Affiliation(s)
- Martina E Spehlmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
28
|
Bacteria in the intestine, helpful residents or enemies from within? Infect Immun 2008; 76:3360-73. [PMID: 18474643 DOI: 10.1128/iai.00187-08] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
29
|
Hodges K, Alto NM, Ramaswamy K, Dudeja PK, Hecht G. The enteropathogenic Escherichia coli effector protein EspF decreases sodium hydrogen exchanger 3 activity. Cell Microbiol 2008; 10:1735-45. [PMID: 18433466 DOI: 10.1111/j.1462-5822.2008.01163.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) have been previously shown to alter sodium hydrogen exchanger 3 (NHE3) activity in human intestinal epithelial cells. To further characterize these observations, PS120 fibroblasts transfected with NHE3 were studied. EPEC E2348/69 infection decreased NHE3 activity in PS120 fibroblasts. The effect on NHE3 was enhanced when PS120 cells were co-transfected with the scaffolding/regulatory proteins NHERF1 or NHERF2 or EBP50 and E3KARP respectively. The decrease in NHE3 activity was dependent on an intact type III secretion system, although intimate attachment mediated by translocated intimin receptor was not required. Despite its ability to bind to NHERF proteins, the EPEC effector Map had no impact on the regulation of NHE activity. Instead, EspF was found to be responsible for decreased NHE3 activity. However, neither EspF-induced apoptosis nor the interaction of EspF with sorting nexin-9, an endocytic protein, were involved.
Collapse
Affiliation(s)
- Kim Hodges
- Section of Digestive Disease and Nutrition, Department Of Medicine, University of Illinois at Chicago, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
30
|
Hodges K, Gill R, Ramaswamy K, Dudeja PK, Hecht G. Rapid activation of Na+/H+ exchange by EPEC is PKC mediated. Am J Physiol Gastrointest Liver Physiol 2006; 291:G959-68. [PMID: 16782696 DOI: 10.1152/ajpgi.00274.2005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) increases sodium/hydrogen exchanger 2 (NHE2)-mediated sodium uptake by intestinal epithelial cells in a type III secretion-dependent manner. However, the mechanism(s) underlying these changes are not known. This study examines the role of a number of known secreted effector molecules and bacterial adhesins as well as the signaling pathways involved in this process. Deletion of the bacterial adhesins Tir and intimin had no effect on the increase in sodium/hydrogen exchanger (NHE) activity promoted by EPEC infection; however, there was a significant decrease upon deletion of the bundle-forming pili. Bacterial supernatant also failed to alter NHE activity, suggesting that direct interaction with bacteria is necessary. Analysis of the signal transduction cascades responsible for the increased NHE2 activity during EPEC infection showed that PLC increased Ca2+, as well as PKCalpha and PKCepsilon were involved in increasing NHE activity. The activation of PKCepsilon by EPEC has not been previously described nor has its role in regulating NHE2 activity. Because EPEC markedly increases NHE2 activity, this pathogen provides an exceptional opportunity to improve our understanding of this less-characterized NHE isoform.
Collapse
Affiliation(s)
- Kim Hodges
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
31
|
Maresca M, Miller D, Quitard S, Dean P, Kenny B. Enteropathogenic Escherichia coli (EPEC) effector-mediated suppression of antimicrobial nitric oxide production in a small intestinal epithelial model system. Cell Microbiol 2006; 7:1749-62. [PMID: 16309461 DOI: 10.1111/j.1462-5822.2005.00587.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In vivo studies with the mouse-specific member of the attaching and effacing (A/E) family of pathogens raised the possibility that these non-invasive enteric pathogens can specifically inhibit inducible nitric oxide synthase (iNOS) expression to prevent the production of antimicrobial nitric oxide (NO). In this study we use polarized Caco-2 cells, a model of human small intestinal epithelia, to (i) demonstrate conclusively that an A/E member, human specific enteropathogenic Escherichia coli (EPEC), can inhibit cytokine-induced iNOS expression, (ii) show that this activity is dependent on the delivery of effector molecules into host cells and (iii) investigate the mechanism of inhibition. Analysis of the level of iNOS-related mRNA, protein and NO production demonstrated that EPEC can inhibit iNOS expression at the transcriptional, by direct and indirect mechanisms, and post-transcriptional levels. This transcriptional block was linked to the failure of the iNOS-related transcriptional factor NF-kappaB, but not STAT1, to undergo phosphorylation-associated activation. A selective pressure to prevent iNOS production was evidenced by the finding that iNOS activity had a potent antimicrobial effect on adherent but not non-adherent bacteria. Moreover, given the central role NF-kappaB plays in transcribing genes associated with early host immune responses, this inhibitory mechanism presumably represents an important role in pathogenesis. Our study also provides insights into the nature of NO production in response to bacterial infection as well as the role of the locus of enterocyte effacement (LEE)-encoded effector molecules in inhibiting iNOS expression.
Collapse
Affiliation(s)
- Marc Maresca
- Institute for Cell and Molecular Biosciences, Medical School, University of Newcastle, Framlington Place, Newcastle upon Tyne, NE24HH, UK
| | | | | | | | | |
Collapse
|
32
|
Shifflett DE, Clayburgh DR, Koutsouris A, Turner JR, Hecht GA. Enteropathogenic E. coli disrupts tight junction barrier function and structure in vivo. J Transl Med 2005; 85:1308-24. [PMID: 16127426 DOI: 10.1038/labinvest.3700330] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) infection disrupts tight junctions (TJs) and perturbs intestinal barrier function in vitro. E. coli secreted protein F (EspF) is, in large part, responsible for these physiological and morphological alterations. We recently reported that the C57BL/6J mouse is a valid in vivo model of EPEC infection as EPEC colonizes the intestinal epithelium and effaces microvilli. Our current aim was to examine the effects of EPEC on TJ structure and barrier function of the mouse intestine and to determine the role of EspF in vivo. C57BL/6J mice were gavaged with approximately 2 x 10(8) EPEC organisms or PBS. At 1 or 5 days postinfection, mice were killed and ileal and colonic tissue was mounted in Ussing chambers to determine barrier function (measured as transepithelial resistance) and short circuit current. TJ structure was analyzed by immunofluorescence microscopy. Wild-type (WT) EPEC significantly diminished the barrier function of ileal and colonic mucosa at 1 and 5 days postinfection. Deficits in barrier function correlated with redistribution of occludin in both tissues. Infection with an EPEC strain deficient of EspF (delta espF) had no effect on barrier function at 1 day postinfection. Furthermore, delta espF had no effect on ileal TJ morphology and minor alterations of colonic TJ morphology at 1 day postinfection. In contrast, at 5 days postinfection, WT EPEC and delta espF had similar effects on barrier function and occludin localization. In both cases this was associated with immune activation, as demonstrated by increased mucosal tumor necrosis factor-alpha levels 5 days postinfection. In conclusion, these data demonstrate that WT EPEC infection of 6-8-week-old C57BL/6J mice (1) significantly decreases barrier function in the ileum and colon (2) redistributes occludin in the ileum and colon and (3) is dependent upon EspF to induce TJ barrier defects at early, but not late, times postinfection.
Collapse
Affiliation(s)
- Donnie E Shifflett
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
33
|
Girard F, Oswald IP, Taranu I, Hélie P, Appleyard GD, Harel J, Fairbrother JM. Host immune status influences the development of attaching and effacing lesions in weaned pigs. Infect Immun 2005; 73:5514-23. [PMID: 16113267 PMCID: PMC1231136 DOI: 10.1128/iai.73.9.5514-5523.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Attaching and effacing Escherichia coli (AEEC) has been associated with naturally occurring attaching and effacing (A/E) lesions in weaned pigs, and although A/E lesions have been experimentally reproduced in newborn piglets, such lesions have been much more difficult to induce in older conventional pigs. Hence, the aim of this study was to examine the effect of oral administration of dexamethasone on the development of A/E lesions in weaned pigs challenged with a porcine enteropathogenic E. coli (PEPEC) strain and to investigate the involvement of local intestinal cytokine response. Dexamethasone, given orally at a dosage of 3 mg kg of body weight(-1), significantly enhanced both the colonization of the challenge strain and the prevalence of foci of intimately adherent bacteria, resulting in extensive A/E lesions in the ileum, cecum, and colon of challenged pigs. We also confirmed the expression of both intimin and Tir by PEPEC strain ECL1001 in A/E lesions in vivo, which is, to our knowledge, the first report of the involvement of the latter proteins in any AEEC infections in vivo. Moreover, semiquantitative reverse transcription-PCR demonstrated that interleukin 1beta (IL-1beta), IL-6, IL-8, and, to a lesser extent, IL-12p40 are significantly upregulated in the ileum following challenge with strain ECL1001, whereas dexamethasone blocks such upregulation. Taken together, our results strongly suggested that host immune status influences the development of A/E lesions in weaned pigs, and it appears that IL-1beta, IL-6, IL-8, and, to a lesser extent, IL-12p40 are expressed during infection of weaned pigs by PEPEC and may contribute to the natural resistance of the host against PEPEC infection.
Collapse
Affiliation(s)
- Francis Girard
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
34
|
Tomson FL, Viswanathan VK, Kanack KJ, Kanteti RP, Straub KV, Menet M, Kaper JB, Hecht G. Enteropathogenic Escherichia coli EspG disrupts microtubules and in conjunction with Orf3 enhances perturbation of the tight junction barrier. Mol Microbiol 2005; 56:447-64. [PMID: 15813736 DOI: 10.1111/j.1365-2958.2005.04571.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
EspG, a secreted effector of enteropathogenic Escherichia coli (EPEC), as well as its homologue Orf3, has been shown to disrupt microtubules (MTs) in fibroblasts and non-polarized epithelial cells. The roles of MTs and the effects of MT disruption in these cell types differ significantly. The aim of this study was to investigate the effects of EspG on polarized, host target intestinal epithelial cells. Immunofluorescent labelling of tubulin showed that EPEC caused progressive fragmentation and loss of the MT network in cells harbouring attached organisms. Immunoblots of proteins extracted from EPEC-infected cells showed a corresponding loss of alpha-tubulin. Type III secretion system (TTSS)-deficient strains had no effect on MT suggesting TTSS dependence. Mutation of espG, but not espF or map, ablated EPEC's effects on MTs for up to 6 h. Ectopic expression of EspG in HeLa cells caused MT disruption. While deletion of espG alone had no effect on the EPEC-induced decrease in transepithelial electrical resistance (TER), mutation of both espG and orf3 significantly delayed the kinetics of this response. Complementation of the double mutant with espG alone restored the kinetics of TER drop to that of wild type. Herein, we describe a previously unrecognized phenotype for the EPEC effectors EspG and Orf3.
Collapse
Affiliation(s)
- Farol L Tomson
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Anselmi L, Cervio E, Guerrini S, Vicini R, Agazzi A, Dellabianca A, Reeve JR, Tonini M, Sternini C. Identification of galanin receptor 1 on excitatory motor neurons in the guinea pig ileum. Neurogastroenterol Motil 2005; 17:273-80. [PMID: 15787947 DOI: 10.1111/j.1365-2982.2004.00590.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Exogenously administered galanin inhibits cholinergic transmission to the longitudinal muscle and reduces peristaltic efficiency in the guinea pig ileum with a mechanism partially mediated by galanin receptor 1 (GAL-R1). We investigated the effect of exogenous galanin 1-16, which has high affinity for GAL-R1, on the ascending excitatory reflex of the circular muscle elicited by radial distension in isolated segments of guinea pig ileum. We used a three-compartment bath that allows dissecting the ascending pathway into the oral (site of excitatory motor neurons), intermediate (site of ascending interneurons) and caudal compartment (site of intrinsic primary afferent neurons). Galanin 1-16 (0.3-3 micromol L(-1)) applied to the oral compartment inhibited in a concentration-dependent manner the ascending excitatory reflex elicited by the wall distension in the caudal compartment. This effect was antagonized by the GAL-R1 antagonist, RWJ-57408 (1 and 10 micromol L(-1)). By contrast, galanin 1-16 was ineffective when added to the intermediate or caudal compartment up to 3 micromol L(-1). GAL-R1 immunoreactive neurons did not contain neuron-specific nuclear protein, a marker for intrinsic primary afferent neurons. These findings indicate that GAL-R1s are present on motor neurons responsible for the ascending excitatory reflex, but not on ascending interneurons and intrinsic primary afferent neurons.
Collapse
Affiliation(s)
- L Anselmi
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Veterans Administration Greater Los Angeles Healthcare System, Bldg. 115, Room 224, Vaglahs, 11301 Wilshire Blvd, Los Angeles, CA 90073, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Savkovic SD, Villanueva J, Turner JR, Matkowskyj KA, Hecht G. Mouse model of enteropathogenic Escherichia coli infection. Infect Immun 2005; 73:1161-70. [PMID: 15664959 PMCID: PMC546940 DOI: 10.1128/iai.73.2.1161-1170.2005] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is an important cause of diarrhea in humans. EPEC infection of cultured intestinal epithelial cells induces attaching and effacing (A/E) lesions, alters intestinal ion transport, increases paracellular permeability, and stimulates inflammation. The lack of a small-animal model has restricted in vivo studies examining EPEC-host interactions. The aim of this study was to characterize the C57BL/6J mouse as a model of EPEC infection. We have shown that EPEC can adhere to and colonize the intestinal epithelium of C57BL/6J mice. Animal weight and water intake were not altered during 10 days of EPEC infection. The proximal colon of infected mice contained semisolid stool, with stool pellets forming only in the distal colon. In contrast, the entire colon of control mice contained formed stool. Microvillous effacement and actin rearrangement, characteristic of A/E lesions, were seen in the intestine of infected mice but not control mice. Histological assessment revealed increased numbers of lamina propria neutrophils with occasional crypt abscesses, intraepithelial lymphocytes, and goblet cells in the intestine of EPEC-infected mice. Altogether, these data suggest that the C57BL/6J mouse is susceptible to infection by EPEC and will provide a suitable in vivo model for studying the consequences of EPEC infection.
Collapse
Affiliation(s)
- Suzana D Savkovic
- Section of Digestive and Liver Diseases, Department of Medicine, University of Illinois, 840 South Wood St., CSB Room 706 (m/c 716), Chicago, IL 60612, USA.
| | | | | | | | | |
Collapse
|
37
|
Tomson FL, Koutsouris A, Viswanathan VK, Turner JR, Savkovic SD, Hecht G. Differing roles of protein kinase C-zeta in disruption of tight junction barrier by enteropathogenic and enterohemorrhagic Escherichia coli. Gastroenterology 2004; 127:859-69. [PMID: 15362041 DOI: 10.1053/j.gastro.2004.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND & AIMS Enteropathogenic Escherichia coli and enterohemorrhagic E. coli harbor highly homologous pathogenicity islands yet show key differences in their mechanisms of action. Both disrupt host intestinal epithelial tight junctions, but the effects of enteropathogenic E. coli are more profound than those of enterohemorrhagic E. coli. The basis for this is not understood. The atypical protein kinase C isoform, protein kinase C-zeta, associates with and regulates the tight junction complex. The aim of this study was to compare the role of protein kinase C-zeta in the disruption of tight junctions after infection with enteropathogenic E. coli and enterohemorrhagic E. coli. METHODS Model intestinal epithelial monolayers infected by enteropathogenic E. coli or enterohemorrhagic E. coli were used for these studies. RESULTS Neither bisindolylmaleimide nor Gö6976, which block several protein kinase C isoforms but not protein kinase C-zeta, protected against the decrease in transepithelial electrical resistance after enteropathogenic E. coli infection. Rottlerin at concentrations that block novel and atypical isoforms, including protein kinase C-zeta, significantly attenuated the decrease in transepithelial electrical resistance. The specific inhibitory peptide, myristoylated protein kinase C-zeta pseudosubstrate, also significantly decreased the enteropathogenic E. coli -associated decrease in transepithelial electrical resistance and redistribution of tight junction proteins. In contrast to enteropathogenic E. coli, the level of protein kinase C-zeta enzyme activity stimulated by enterohemorrhagic E. coli was transient and minor, and protein kinase C-zeta inhibition had no effect on the decrease in transepithelial electrical resistance or the redistribution of occludin. CONCLUSIONS The differential regulation of protein kinase C-zeta by enteropathogenic E. coli and enterohemorrhagic E. coli may in part explain the less profound effect of the latter on the barrier function of tight junctions.
Collapse
Affiliation(s)
- Farol L Tomson
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, Chicago, USA
| | | | | | | | | | | |
Collapse
|
38
|
Hecht G, Hodges K, Gill RK, Kear F, Tyagi S, Malakooti J, Ramaswamy K, Dudeja PK. Differential regulation of Na+/H+ exchange isoform activities by enteropathogenic E. coli in human intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2004; 287:G370-8. [PMID: 15075254 DOI: 10.1152/ajpgi.00432.2003] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) is an important human intestinal foodborne pathogen associated with diarrhea, especially in infants and young children. Although EPEC produces characteristic attaching and effacing lesions and loss of microvilli, the pathophysiology of EPEC-associated diarrhea, particularly during early infection, remains elusive. The present studies were designed to examine the direct effects of EPEC infection on intestinal absorption via Na(+)/H(+) exchanger (NHE) isoforms. Caco-2 cells were infected with EPEC strain E2348/69 or nonpathogenic E. coli HB101 for a period of 60 to 120 min. Total NHE activity was significantly increased at 60 min, reaching approximately threefold increase after 90 min of EPEC infection. Similar findings were seen in HT-29 cells and T84 cells indicating that the response was not cell-line specific. Most surprising was the differential regulation of NHE2 and NHE3 by EPEC. Marked activation of NHE2 (300%) occurred, whereas significant inhibition ( approximately 50%) of NHE3 activity was induced. The activity of basolateral isoform NHE1 was also significantly increased in response to EPEC infection. Mutations that disrupted the type III secretion system (TTSS) ablated the effect of EPEC on the activity of both NHE2 and NHE3. These results suggest that EPEC, through a TTSS-dependent mechanism, exerts differential effects on NHE isoform activity in intestinal epithelial cells. Additionally, NHEs do not appear to play any role in EPEC-mediated inflammation, because the NHE inhibitors amiloride and 5-(N-ethyl-N-isopropyl)amiloride did not prevent EPEC-mediated IkappaBalpha degradation.
Collapse
Affiliation(s)
- Gail Hecht
- Univ. of Illinois at Chicago, Medical Research Service (600/151 Chicago Veterans Affairs Health Care System, 820 S. Damen Ave., Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Matkowskyj KA, Nathaniel R, Prasad R, Weihrauch D, Rao M, Benya RV. Galanin contributes to the excess colonic fluid secretion observed in dextran sulfate sodium murine colitis. Inflamm Bowel Dis 2004; 10:408-16. [PMID: 15475749 DOI: 10.1097/00054725-200407000-00012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Galanin is present in enteric nerves lining the gastrointestinal (GI) tract where it is normally involved in regulating intestinal motility by binding to the galanin-1 receptor (Gal1R) subtype expressed by smooth muscle cells. In contrast, although epithelial cells lining the colon do not normally express Gal1R, this protein is up-regulated by the inflammation-associated transcription factor NF-kappaB. We previously showed that the murine colitis induced by dextran sulfate sodium (DSS) was associated with increased Gal1R expression as well as by increased colonic fluid secretion. Although Gal1R up-regulation by colonic epithelial cells results in increased intestinal Cl- secretion, the relative contributions of galanin to this excess colonic fluid secretion could not be determined. We therefore created a mouse genetically incapable of synthesizing Gal1R (GAL1R-/- mice). We herein demonstrate that both wild-type and GAL1R-/- mice developed identical histologic lesions in response to DSS. This was characterized by a marked inflammatory infiltrate, activation of NF-kappaB in both enterocytes and enteric nerves, and a threefold increase in neuronal galanin. Colonic fluid secretion, while increased, was approximately half that in GAL1R-/- mice as compared with their wild-type littermates. Overall, then, these findings strongly suggest that approximately half of the increase in colonic fluid secretion in DSS colitis is due to up-regulation of the Gal1R.
Collapse
Affiliation(s)
- Kristina A Matkowskyj
- Departments of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Few microorganisms are as versatile as Escherichia coli. An important member of the normal intestinal microflora of humans and other mammals, E. coli has also been widely exploited as a cloning host in recombinant DNA technology. But E. coli is more than just a laboratory workhorse or harmless intestinal inhabitant; it can also be a highly versatile, and frequently deadly, pathogen. Several different E. coli strains cause diverse intestinal and extraintestinal diseases by means of virulence factors that affect a wide range of cellular processes.
Collapse
Affiliation(s)
- James B Kaper
- Center for Vaccine Development, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | | | |
Collapse
|
41
|
Shen PJ, Larm JA, Gundlach AL. Expression and plasticity of galanin systems in cortical neurons, oligodendrocyte progenitors and proliferative zones in normal brain and after spreading depression. Eur J Neurosci 2003; 18:1362-76. [PMID: 14511317 DOI: 10.1046/j.1460-9568.2003.02860.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neocortex contains very few galanin neurons but receives a moderate galanin innervation from various subcortical loci. Recent data suggest that galanin helps regulate the tonic neuronal excitability of hippocampus and probably cerebral cortex but relatively little is known about the anatomy and functional regulation of cortical galanin systems. Therefore, we examined, in the rat, the effect of the intense but benign stimulus, cortical spreading depression (CSD), on the expression of galanin and galanin receptors (GalR1 and GalR2) in the neocortex and associated regions, revealing complex, multicellular responses. Thus, following acute, unilateral KCl-induced CSD, a delayed and transient induction (onset after 48 h, lasting approximately 24 h) of galanin mRNA and peptide production occurred across the ipsilateral cerebral cortex in activated oligodendrocyte progenitor cells (OPCs), identified by specific NG2 proteoglycan immunostaining. An increase in GalR1 mRNA, immunoreactivity and receptor binding occurred in neurons within layers II and V of neocortex and in piriform cortex at 7-28 days after CSD, associated with a long-lasting depletion of galanin-positive nerve fibres in these regions. In contrast, GalR2 mRNA expression was largely unaltered after CSD. Additional novel findings in normal, adult brain were the detection of galanin mRNA and immunoreactivity in OPCs within the medial corpus callosum and in immature progenitor cells in the subventricular zone and rostral migratory stream. GalR1 and GalR2 mRNA was also present in these latter regions. These findings and the complex modulation of galanin and galanin receptors in multiple cell types (neurons/OPCs) following acute cortical activation/depression further demonstrate the potential plasticity of neuronal and non-neuronal galanin systems under physiological and pathological conditions and strongly suggest additional functions for this pleiotropic peptide in mammalian brain.
Collapse
Affiliation(s)
- Pei-Juan Shen
- Howard Florey Institute of Experimental Physiology and Medicine, Austin and Repatriation Medical Centre, The University of Melbourne, Victoria 3010, Australia
| | | | | |
Collapse
|
42
|
Resta-Lenert S, Barrett KE. Live probiotics protect intestinal epithelial cells from the effects of infection with enteroinvasive Escherichia coli (EIEC). Gut 2003; 52:988-97. [PMID: 12801956 PMCID: PMC1773702 DOI: 10.1136/gut.52.7.988] [Citation(s) in RCA: 417] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2003] [Indexed: 12/17/2022]
Abstract
BACKGROUND The colonic epithelium maintains a life long reciprocally beneficial interaction with the colonic microbiota. Disruption is associated with mucosal injury. AIMS We hypothesised that probiotics may limit epithelial damage induced by enteroinvasive pathogens, and promote restitution. METHODS Human intestinal epithelial cell lines (HT29/cl.19A and Caco-2) were exposed to enteroinvasive Escherichia coli (EIEC 029:NM), and/or probiotics (Streptococcus thermophilus (ST), ATCC19258, and Lactobacillus acidophilus (LA), ATCC4356). Infected cells and controls were assessed for transepithelial resistance, chloride secretory responses, alterations in cytoskeletal and tight junctional proteins, and responses to epidermal growth factor (EGF) stimulation. RESULTS Exposure of cell monolayers to live ST/LA, but not to heat inactivated ST/LA, significantly limited adhesion, invasion, and physiological dysfunction induced by EIEC. Antibiotic killed ST/LA reduced adhesion somewhat but were less effective in limiting the consequences of EIEC invasion of cell monolayers. Furthermore, live ST/LA alone increased transepithelial resistance, contrasting markedly with the fall in resistance evoked by EIEC infection, which could also be blocked by live ST/LA. The effect of ST/LA on resistance was accompanied by maintenance (actin, ZO-1) or enhancement (actinin, occludin) of cytoskeletal and tight junctional protein phosphorylation. ST/LA had no effect on chloride secretion by themselves but reversed the increase in basal secretion evoked by EIEC. EIEC also reduced the ability of EGF to activate its receptor, which was reversed by ST/LA. CONCLUSIONS Live ST/LA interact with intestinal epithelial cells to protect them from the deleterious effect of EIEC via mechanisms that include, but are not limited to, interference with pathogen adhesion and invasion. Probiotics likely also enhance the barrier function of naïve epithelial cells not exposed to any pathogen.
Collapse
Affiliation(s)
- S Resta-Lenert
- Department of Medicine, School of Medicine, University of California-San Diego, UCSD Medical Center 8414, 200 West Arbor Drive, San Diego, CA 92103, USA.
| | | |
Collapse
|
43
|
Berkes J, Viswanathan VK, Savkovic SD, Hecht G. Intestinal epithelial responses to enteric pathogens: effects on the tight junction barrier, ion transport, and inflammation. Gut 2003; 52:439-51. [PMID: 12584232 PMCID: PMC1773546 DOI: 10.1136/gut.52.3.439] [Citation(s) in RCA: 435] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The effects of pathogenic organisms on host intestinal epithelial cells are vast. Innumerable signalling pathways are triggered leading ultimately to drastic changes in physiological functions. Here, the ways in which enteric bacterial pathogens utilise and impact on the three major physiological functions of the intestinal epithelium are discussed: alterations in the structure and function of the tight junction barrier, induction of fluid and electrolyte secretion, and activation of the inflammatory cascade. This field of investigation, which was virtually non-existent a decade ago, has now exploded, thus rapidly expanding our understanding of bacterial pathogenesis. Through increased delineation of the ways in which microbes alter host physiology, we simultaneous gain insight into the normal regulatory mechanisms of the intestinal epithelium.
Collapse
Affiliation(s)
- J Berkes
- Section of Digestive Diseases and Nutrition, University of Illinois at Chicago and Chicago Veterans Administration Medical Center, West Side Division, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
44
|
Hauf N, Chakraborty T. Suppression of NF-kappa B activation and proinflammatory cytokine expression by Shiga toxin-producing Escherichia coli. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:2074-82. [PMID: 12574378 DOI: 10.4049/jimmunol.170.4.2074] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The NF-kappaB family of transcription factors forms one of the first lines of defense against infectious disease by inducing the expression of genes involved in inflammatory and immune responses. In this study, we analyzed the impact of Shiga toxin-producing Escherichia coli (STEC) on the NF-kappaB DNA-binding activity in HeLa cells. After a period of weak initial activation, DNA binding of NF-kappaB was actively suppressed by viable, E. coli secreted protein B (EspB)-secreting STEC. Sustained NF-kappaB activity was observed either using an isogenic mutant lacking EspB or after gentamicin-based killing of STEC after allowing bacterial attachment. These observations indicate that the ability of STEC to cause NF-kappaB activation is suppressed by a translocated bacterial effector protein, which is either EspB itself or requires EspB for delivery into the host cell. We found that STEC, enterohemorrhagic E. coli, and enteropathogenic E. coli all interfere with NF-kappaB activation initiated by TNF-alpha, indicating that suppression of signal-induced NF-kappaB activity is a property common to several attaching and effacing bacteria. As a consequence of NF-kappaB suppression, wild-type STEC induces significantly lower mRNA levels of IL-8, IL-6, and IL-1alpha upon prolonged infection periods compared with bacteria lacking EspB. For IL-8 and IL-6, the suppressive effect was also reflected at the level of cytokine secretion. Suppression of both basal and signal-induced NF-kappaB DNA binding by attaching and effacing-inducing bacteria appears to be an active strategy to counteract host defense responses, thus favoring intestinal colonization by these pathogens.
Collapse
Affiliation(s)
- Nadja Hauf
- Institut für Medizinische Mikrobiologie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 107, 35392 Giessen, Germany
| | | |
Collapse
|
45
|
Larm JA, Shen PJ, Gundlach AL. Differential galanin receptor-1 and galanin expression by 5-HT neurons in dorsal raphé nucleus of rat and mouse: evidence for species-dependent modulation of serotonin transmission. Eur J Neurosci 2003; 17:481-93. [PMID: 12581166 DOI: 10.1046/j.1460-9568.2003.02471.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Galanin and galanin receptors are widely expressed by neurons in rat brain that either synthesize/release and/or are responsive to, classical transmitters such as gamma-aminobutyric acid, acetylcholine, noradrenaline, histamine, dopamine and serotonin (5-hydroxytryptamine, 5-HT). The dorsal raphé nucleus (DRN) contains approximately 50% of the 5-HT neurons in the rat brain and a high percentage of these cells coexpress galanin and are responsive to exogenous galanin in vitro. However, the precise identity of the galanin receptor(s) present on these 5-HT neurons has not been previously established. Thus, the current study used a polyclonal antibody for the galanin receptor-1 (GalR1) to examine the possible expression of this receptor within the DRN of the rat and for comparative purposes also in the mouse. In the rat, intense GalR1-immunoreactivity (IR) was detected in a substantial population of 5-HT-immunoreactive neurons in the DRN, with prominent receptor immunostaining associated with soma and proximal dendrites. GalR1-IR was also observed in many cells within the adjacent median raphé nucleus. In mouse DRN, neurons exhibited similar levels and distribution of 5-HT-IR to that in the rat, but GalR1-IR was undetectable. Consistent with this, galanin and GalR1 mRNA were also undetectable in mouse DRN by in situ hybridization histochemistry, despite the detection of GalR1 mRNA (and GalR1-IR) in adjacent cells in the periaqueductal grey and other midbrain areas. 5-HT neuron activity in the DRN is primarily regulated via 5-HT1A autoreceptors, via inhibition of adenylate cyclase and activation of inward-rectifying K+ channels. Notably, the GalR1 receptor subtype signals via identical mechanisms and our findings establish that galanin modulates 5-HT neuron activity in the DRN of the rat via GalR1 (auto)receptors. However, these studies also identify important species differences in the relationship between midbrain galanin and 5-HT systems, which should prompt further investigations in relation to comparative human neurochemistry and which have implications for studies of animal models of relevant neurological conditions such as stress, anxiety and depression.
Collapse
Affiliation(s)
- Jari A Larm
- Howard Florey Institute of Experimental Physiology and Medicine, Austin & Repatriation Medical Centre, The University of Melbourne, Victoria, Australia
| | | | | |
Collapse
|
46
|
Saban MR, Nguyen NB, Hammond TG, Saban R. Gene expression profiling of mouse bladder inflammatory responses to LPS, substance P, and antigen-stimulation. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:2095-110. [PMID: 12057914 PMCID: PMC1850812 DOI: 10.1016/s0002-9440(10)61159-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Inflammatory bladder disorders such as interstitial cystitis (IC) deserve attention since a major problem of the disease is diagnosis. IC affects millions of women and is characterized by severe pain, increased frequency of micturition, and chronic inflammation. Characterizing the molecular fingerprint (gene profile) of IC will help elucidate the mechanisms involved and suggest further approaches for therapeutic intervention. Therefore, in the present study we used established animal models of cystitis to determine the time course of bladder inflammatory responses to antigen, Escherichia coli lipopolysaccharide (LPS), and substance P (SP) by morphological analysis and cDNA microarrays. The specific aim of the present study was to compare bladder inflammatory responses to antigen, LPS, and SP by morphological analysis and cDNA microarray profiling to determine whether bladder responses to inflammation elicit a specific universal gene expression response regardless of the stimulating agent. During acute bladder inflammation, there was a predominant infiltrate of polymorphonuclear neutrophils into the bladder. Time-course studies identified early, intermediate, and late genes that were commonly up-regulated by all three stimuli. These genes included: phosphodiesterase 1C, cAMP-dependent protein kinase, iNOS, beta-NGF, proenkephalin B and orphanin, corticotrophin-releasing factor (CRF) R, estrogen R, PAI2, and protease inhibitor 17, NFkB p105, c-fos, fos-B, basic transcription factors, and cytoskeleton and motility proteins. Another cluster indicated genes that were commonly down-regulated by all three stimuli and included HSF2, NF-kappa B p65, ICE, IGF-II and FGF-7, MMP2, MMP14, and presenilin 2. Furthermore, we determined gene profiles that identify the transition between acute and chronic inflammation. During chronic inflammation, the urinary bladder presented a predominance of monocyte/macrophage infiltrate and a concomitant increase in the expression of the following genes: 5-HT 1c, 5-HTR7, beta 2 adrenergic receptor, c-Fgr, collagen 10 alpha 1, mast cell factor, melanocyte-specific gene 2, neural cell adhesion molecule 2, potassium inwardly-rectifying channel, prostaglandin F receptor, and RXR-beta cis-11-retinoic acid receptor. We conclude that microarray analysis of genes expressed in the bladder during experimental inflammation may be predictive of outcome. Further characterization of the inflammation-induced gene expression profiles obtained here may identify novel biomarkers and shed light into the etiology of cystitis.
Collapse
Affiliation(s)
- Marcia R Saban
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | |
Collapse
|
47
|
Jones SL, Blikslager AT. Role of the Enteric Nervous System in the Pathophysiology of Secretory Diarrhea. J Vet Intern Med 2002. [DOI: 10.1111/j.1939-1676.2002.tb02361.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
48
|
Resta-Lenert S, Barrett KE. Enteroinvasive bacteria alter barrier and transport properties of human intestinal epithelium: role of iNOS and COX-2. Gastroenterology 2002; 122:1070-87. [PMID: 11910358 DOI: 10.1053/gast.2002.32372] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Various invasive pathogens cause diarrhea, but the mechanism(s) are poorly understood. We hypothesized that nitric oxide and prostaglandins might modulate chloride secretory and barrier properties of the infected intestinal epithelium and that diarrhea is caused, in part, by altered expression of inducible NO synthase (iNOS) and cyclooxygenase 2 (COX-2). METHODS Studies were conducted in human intestinal epithelial cell lines (HT29/cl.19A, Caco-2, and T84). Cells were infected with enteroinvasive Escherichia coli (EIEC 029:NM) or Salmonella dublin (SD), or nonpathogenic, noninvasive bacteria (Streptococcus thermophilus [ST] and Lactobacillus acidophilus [LA]). Infected cells and controls were tested for transepithelial resistance, chloride secretion, prostaglandin E2, guanosine 3',5'-cyclic monophosphate and adenosine 3',5'-cyclic monophosphate, and protein expression. RESULTS Cells infected with EIEC or SD, but not uninfected controls or ST/LA-exposed monolayers, showed a progressive reduction in transepithelial resistance starting at 6-12 hours. Infected HT29/cl.19A and Caco-2 cells, but not T84 cells, also showed an increase in total nitrite. Expression of iNOS, and consequently COX-2, was also increased, followed by increased production of prostaglandins and cyclic nucleotides. Furthermore, basal and stimulated chloride secretory responses to various agonists were enhanced in HT29/cl.19A and Caco-2 cells after infection with enteroinvasive bacteria, and this effect was reversed for some agonists by iNOS or COX-2 inhibitors. Increased expression of cystic fibrosis transmembrane conductance regulator and NKCC1 was also observed in EIEC or SD-infected cells vs. controls, secondary to NO synthase activity. CONCLUSIONS Up-regulation of iNOS and COX-2 by enteroinvasive bacteria can modulate chloride secretion and barrier function in intestinal epithelial cells. Thus, these enzymes represent possible therapeutic targets in infectious diarrhea.
Collapse
Affiliation(s)
- Silvia Resta-Lenert
- Department of Medicine, and Biomedical Sciences Ph.D. Program, University of California, San Diego, School of Medicine, San Diego, California, USA.
| | | |
Collapse
|
49
|
Abstract
Many pathogens must surmount an epithelial cell barrier in order to establish an infection. While much has been learned about the interaction of bacterial pathogens with cultured epithelial cells, the influence of cell polarity on these events has only recently been appreciated. This review outlines bacterial-host epithelial cell interactions in the context of the distinct apical and basolateral surfaces of the polarized epithelium that lines the lumens of our organs.
Collapse
Affiliation(s)
- B I Kazmierczak
- Department of Medicine, University of California, San Francisco, California 94143-0654, USA.
| | | | | |
Collapse
|
50
|
Savkovic SD, Ramaswamy A, Koutsouris A, Hecht G. EPEC-activated ERK1/2 participate in inflammatory response but not tight junction barrier disruption. Am J Physiol Gastrointest Liver Physiol 2001; 281:G890-8. [PMID: 11557508 DOI: 10.1152/ajpgi.2001.281.4.g890] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC) alters many functions of the host intestinal epithelia. Inflammation is initiated by activation of nuclear factor (NF)-kappaB, and paracellular permeability is enhanced via a Ca2+- and myosin light-chain kinase (MLCK)-dependent pathway. The aims of this study were to identify signaling pathways by which EPEC triggers inflammation and to determine whether these pathways parallel or diverge from those that alter permeability. EPEC-induced phosphorylation and degradation of the primary inhibitor of NF-kappaB (IkappaBalpha) were tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta independent. In contrast to Salmonella typhimurium, EPEC-stimulated IkappaBalpha degradation and IL-8 expression did not require Ca2+. Instead, extracellular signal-regulated kinase (ERK)-1/2 was significantly and rapidly activated. ERK1/2 inhibitors attenuated IkappaBalpha degradation and IL-8 expression. Although ERK1/2 can activate MLCK, its inhibition had no impact on EPEC disruption of the tight junction barrier. In conclusion, EPEC-induced inflammation 1) is TNF-alpha and IL-1beta receptor independent, 2) utilizes pathways differently from S. typhimurium, 3) requires ERK1/2, and 4) employs signals that are distinct from those that alter permeability. This is the first time that EPEC-activated signaling cascades have been linked to independent functional consequences.
Collapse
Affiliation(s)
- S D Savkovic
- Department of Medicine, Section of Digestive and Liver Diseases, University of Illinois, West Side Veterans Affairs Medical Center, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|