1
|
Arévalo-Jaimes BV, Salinas-Pena M, Ponte I, Jordan A, Roque A, Torrents E. Antimicrobial and antibiofilm activity of human recombinant H1 histones against bacterial infections. mSystems 2024:e0070424. [PMID: 39470247 DOI: 10.1128/msystems.00704-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Histones possess significant antimicrobial potential, yet their activity against biofilms remains underexplored. Moreover, concerns regarding adverse effects limit their clinical implementation. We investigated the antibacterial efficacy of human recombinant histone H1 subtypes against Pseudomonas aeruginosa PAO1, both planktonic and in biofilms. After the in vitro tests, toxicity and efficacy were assessed in a P. aeruginosa PAO1 infection model using Galleria mellonella larvae. Histones were also evaluated in combination with ciprofloxacin (Cpx) and gentamicin (Gm). Our results demonstrate antimicrobial activity of all three histones against P. aeruginosa PAO1, with H1.0 and H1.4 showing efficacy at lower concentrations. The bactericidal effect was associated with a mechanism of membrane disruption. In vitro studies using static and dynamic models showed that H1.4 had antibiofilm potential by reducing cell biomass. Neither H1.0 nor H1.4 showed toxicity in G. mellonella larvae, and both increased larvae survival when infected with P. aeruginosa PAO1. Although in vitro synergism was observed between ciprofloxacin and H1.0, no improvement over the antibiotic alone was noted in vivo. Differences in antibacterial and antibiofilm activity were attributed to sequence and structural variations among histone subtypes. Moreover, the efficacy of H1.0 and H1.4 was influenced by the presence and strength of the extracellular matrix. These findings suggest histones hold promise for combating acute and chronic infections caused by pathogens such as P. aeruginosa.IMPORTANCEThe constant increase of multidrug-resistant bacteria is a critical global concern. The inefficacy of current therapies to treat bacterial infections is attributed to multiple mechanisms of resistance, including the capacity to form biofilms. Therefore, the identification of novel and safe therapeutic strategies is imperative. This study confirms the antimicrobial potential of three histone H1 subtypes against both Gram-negative and Gram-positive bacteria. Furthermore, histones H1.0 and H1.4 demonstrated in vivo efficacy without associated toxicity in an acute infection model of Pseudomonas aeruginosa PAO1 in Galleria mellonella larvae. The bactericidal effect of these proteins also resulted in biomass reduction of P. aeruginosa PAO1 biofilms. Given the clinical significance of this opportunistic pathogen, our research provides a comprehensive initial evaluation of the efficacy, toxicity, and mechanism of action of a potential new therapeutic approach against acute and chronic bacterial infections.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | - Inmaculada Ponte
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alicia Roque
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Ljungholm PL, Ermund A, Söderlund Garsveden MM, Pettersson VL, Gustafsson JK. The anion exchanger slc26a3 regulates colonic mucus expansion during steady state and in response to prostaglandin E 2, while Cftr regulates de novo mucus release in response to carbamylcholine. Pflugers Arch 2024; 476:1209-1219. [PMID: 38829391 PMCID: PMC11271379 DOI: 10.1007/s00424-024-02975-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/05/2024]
Abstract
The intestinal epithelium is covered by mucus that protects the tissue from the luminal content. Studies have shown that anion secretion via the cystic fibrosis conductance regulator (Cftr) regulates mucus formation in the small intestine. However, mechanisms regulating mucus formation in the colon are less understood. The aim of this study was to explore the role of anion transport in the regulation of mucus formation during steady state and in response to carbamylcholine (CCh) and prostaglandin E2 (PGE2). The broad-spectrum anion transport inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonate (DIDS), CftrdF508 (CF) mice, and the slc26a3 inhibitor SLC26A3-IN-2 were used to inhibit anion transport. In the distal colon, steady-state mucus expansion was reduced by SLC26A3-IN-2 and normal in CF mice. PGE2 stimulated mucus expansion without de novo mucus release in wild type (WT) and CF colon via slc26a3 sensitive mechanisms, while CCh induced de novo mucus secretion in WT but not in CF colon. However, when added simultaneously, CCh and PGE2 stimulated de novo mucus secretion in the CF colon via DIDS-sensitive pathways. A similar response was observed in CF ileum that responded to CCh and PGE2 with DIDS-sensitive de novo mucus secretion. In conclusion, this study suggests that slc26a3 regulates colonic mucus expansion, while Cftr regulates CCh-induced de novo mucus secretion from ileal and distal colon crypts. Furthermore, these findings demonstrate that in the absence of a functional Cftr channel, parallel stimulation with CCh and PGE2 activates additional anion transport processes that help release mucus from intestinal goblet cells.
Collapse
Affiliation(s)
- Penny L Ljungholm
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Victor L Pettersson
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden
| | - Jenny K Gustafsson
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden.
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
3
|
Kassai S, de Vos P. Gastrointestinal barrier function, immunity, and neurocognition: The role of human milk oligosaccharide (hMO) supplementation in infant formula. Compr Rev Food Sci Food Saf 2024; 23:e13271. [PMID: 38284595 DOI: 10.1111/1541-4337.13271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/06/2023] [Accepted: 10/27/2023] [Indexed: 01/30/2024]
Abstract
Breastmilk is seen as the gold standard for infant nutrition as it provides nutrients and compounds that stimulate gut barrier, immune, and brain development to the infant. However, there are many instances where it is not possible for an infant to be fed with breastmilk, especially for the full 6 months recommended by the World Health Organization. In such instances, infant formula is seen as the next best approach. However, infant formulas do not contain human milk oligosaccharides (hMOs), which are uniquely present in human milk as the third most abundant solid component. hMOs have been linked to many health benefits, such as the development of the gut microbiome, the immune system, the intestinal barrier, and a healthy brain. This paper reviews the effects of specific hMOs applied in infant formula on the intestinal barrier, including the not-often-recognized intestinal alkaline phosphatase system that prevents inflammation. Additionally, impact on immunity and the current proof for effects in neurocognitive function and the corresponding mechanisms are discussed. Recent studies suggest that hMOs can alter gut microbiota, modulate intestinal immune barrier function, and promote neurocognitive function. The hMOs 2'-fucosyllactose and lacto-N-neotetraose have been found to have positive effects on the development of infants and have been deemed safe for use in formula. However, their use has been limited due to their cost and complexity of synthesis. Thus, although many benefits have been described, complex hMOs and combinations of hMOs with other oligosaccharides are the best approach to stimulate gut barrier, immune, and brain development and for the prevention of disease.
Collapse
Affiliation(s)
- Sonia Kassai
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Kim DK, Han D, Bae J, Kim H, Lee S, Kim JS, Jeong YG, Shin J, Park HW. Verapamil-loaded supramolecular hydrogel patch attenuates metabolic dysfunction-associated fatty liver disease via restoration of autophagic clearance of aggregated proteins and inhibition of NLRP3. Biomater Res 2023; 27:4. [PMID: 36670488 PMCID: PMC9854054 DOI: 10.1186/s40824-023-00342-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/15/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Obesity, a serious threat to public health, is linked to chronic metabolic complications including insulin resistance, type-2 diabetes, and metabolic dysfunction-associated fatty liver disease (MAFLD). Current obesity medications are challenged by poor effectiveness, poor patient compliance, and potential side effects. Verapamil is an inhibitor of L-type calcium channels, FDA-approved for the treatment of hypertension. We previously investigated the effect of verapamil on modulating autophagy to treat obesity-associated lipotoxicity. This study aims to develop a verapamil transdermal patch and to evaluate its anti-obesity effects. METHODS Verapamil is loaded in biomimetic vascular bundle-like carboxymethyl pullulan-based supramolecular hydrogel patches cross-linked with citric acid and glycerol linkages (CLCMP). The investigation was then carried out to determine the therapeutic effect of verapamil-loaded CLCMP (Vera@CLCMP) on diet-induced obese mice. RESULTS Vera@CLCMP hydrogel patches with hierarchically organized and anisotropic pore structures not only improved verapamil bioavailability without modifying its chemical structure but also enhanced verapamil release through the stratum corneum barrier. Vera@CLCMP patches exhibit low toxicity and high effectiveness at delivering verapamil into the systemic circulation through the dermis in a sustained manner. Specifically, transdermal administration of this patch into diet-induced obese mice drastically improved glucose tolerance and insulin sensitivity and alleviated metabolic derangements associated with MAFLD. Furthermore, we uncovered a distinct molecular mechanism underlying the anti-obesity effects associated with the hepatic NLR family pyrin domain-containing 3 (NLRP3) inflammasome and autophagic clearance by the vera@CLCMP hydrogel patches. CONCLUSION The current study provides promising drug delivery platforms for long-term family treatment of chronic diseases, including obesity and metabolic dysfunctions.
Collapse
Affiliation(s)
- Do Kyung Kim
- grid.411143.20000 0000 8674 9741Department of Anatomy, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Daewon Han
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Jeongyun Bae
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Haeil Kim
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Solji Lee
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Jong-Seok Kim
- grid.411143.20000 0000 8674 9741Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Young-Gil Jeong
- grid.411143.20000 0000 8674 9741Department of Anatomy, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Jongdae Shin
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea ,grid.411143.20000 0000 8674 9741Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| | - Hwan-Woo Park
- grid.411143.20000 0000 8674 9741Department of Cell Biology, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea ,grid.411143.20000 0000 8674 9741Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, 35365 Republic of Korea
| |
Collapse
|
5
|
Yin Y, Guo Q, Zhou X, Duan Y, Yang Y, Gong S, Han M, Liu Y, Yang Z, Chen Q, Li F. Role of brain-gut-muscle axis in human health and energy homeostasis. Front Nutr 2022; 9:947033. [PMID: 36276808 PMCID: PMC9582522 DOI: 10.3389/fnut.2022.947033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
The interrelationship between brain, gut and skeletal muscle plays a key role in energy homeostasis of the body, and is becoming a hot topic of research. Intestinal microbial metabolites, such as short-chain fatty acids (SCFAs), bile acids (BAs) and tryptophan metabolites, communicate with the central nervous system (CNS) by binding to their receptors. In fact, there is a cross-talk between the CNS and the gut. The CNS, under the stimulation of pressure, will also affect the stability of the intestinal system, including the local intestinal transport, secretion and permeability of the intestinal system. After the gastrointestinal tract collects information about food absorption, it sends signals to the central system through vagus nerve and other channels to stimulate the secretion of brain-gut peptide and produce feeding behavior, which is also an important part of maintaining energy homeostasis. Skeletal muscle has receptors for SCFAs and BAs. Therefore, intestinal microbiota can participate in skeletal muscle energy metabolism and muscle fiber conversion through their metabolites. Skeletal muscles can also communicate with the gut system during exercise. Under the stimulation of exercise, myokines secreted by skeletal muscle causes the secretion of intestinal hormones, and these hormones can act on the central system and affect food intake. The idea of the brain-gut-muscle axis is gradually being confirmed, and at present it is important for regulating energy homeostasis, which also seems to be relevant to human health. This article focuses on the interaction of intestinal microbiota, central nervous, skeletal muscle energy metabolism, and feeding behavior regulation, which will provide new insight into the diagnostic and treatment strategies for obesity, diabetes, and other metabolic diseases.
Collapse
Affiliation(s)
- Yunju Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Xihong Zhou
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Saiming Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Mengmeng Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yating Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhikang Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Qinghua Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Transcriptional Profiling of the Small Intestine and the Colon Reveals Modulation of Gut Infection with Citrobacter rodentium According to the Vitamin A Status. Nutrients 2022; 14:nu14081563. [PMID: 35458125 PMCID: PMC9026425 DOI: 10.3390/nu14081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022] Open
Abstract
Vitamin A (VA) deficiency and diarrheal diseases are both serious public health issues worldwide. VA deficiency is associated with impaired intestinal barrier function and increased risk of mucosal infection-related mortality. The bioactive form of VA, retinoic acid, is a well-known regulator of mucosal integrity. Using Citrobacter rodentium-infected mice as a model for diarrheal diseases in humans, previous studies showed that VA-deficient (VAD) mice failed to clear C. rodentium as compared to their VA-sufficient (VAS) counterparts. However, the distinct intestinal gene responses that are dependent on the host’s VA status still need to be discovered. The mRNAs extracted from the small intestine (SI) and the colon were sequenced and analyzed on three levels: differential gene expression, enrichment, and co-expression. C. rodentium infection interacted differentially with VA status to alter colon gene expression. Novel functional categories downregulated by this pathogen were identified, highlighted by genes related to the metabolism of VA, vitamin D, and ion transport, including improper upregulation of Cl− secretion and disrupted HCO3− metabolism. Our results suggest that derangement of micronutrient metabolism and ion transport, together with the compromised immune responses in VAD hosts, may be responsible for the higher mortality to C. rodentium under conditions of inadequate VA.
Collapse
|
7
|
Immunomodulatory and Allergenic Properties of Antimicrobial Peptides. Int J Mol Sci 2022; 23:ijms23052499. [PMID: 35269641 PMCID: PMC8910669 DOI: 10.3390/ijms23052499] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
With the growing problem of the emergence of antibiotic-resistant bacteria, the search for alternative ways to combat bacterial infections is extremely urgent. While analyzing the effect of antimicrobial peptides (AMPs) on immunocompetent cells, their effect on all parts of the immune system, and on humoral and cellular immunity, is revealed. AMPs have direct effects on neutrophils, monocytes, dendritic cells, T-lymphocytes, and mast cells, participating in innate immunity. They act on B-lymphocytes indirectly, enhancing the induction of antigen-specific immunity, which ultimately leads to the activation of adaptive immunity. The adjuvant activity of AMPs in relation to bacterial and viral antigens was the reason for their inclusion in vaccines and made it possible to formulate the concept of a “defensin vaccine” as an innovative basis for constructing vaccines. The immunomodulatory function of AMPs involves their influence on cells in the nearest microenvironment, recruitment and activation of other cells, supporting the response to pathogenic microorganisms and completing the inflammatory process, thus exhibiting a systemic effect. For the successful use of AMPs in medical practice, it is necessary to study their immunomodulatory activity in detail, taking into account their pleiotropy. The degree of maturity of the immune system and microenvironment can contribute to the prevention of complications and increase the effectiveness of therapy, since AMPs can suppress inflammation in some circumstances, but aggravate the response and damage of organism in others. It should also be taken into account that the real functions of one or another AMP depend on the types of total regulatory effects on the target cell, and not only on properties of an individual peptide. A wide spectrum of biological activity, including direct effects on pathogens, inactivation of bacterial toxins and influence on immunocompetent cells, has attracted the attention of researchers, however, the cytostatic activity of AMPs against normal cells, as well as their allergenic properties and low stability to host proteases, are serious limitations for the medical use of AMPs. In this connection, the tasks of searching for compounds that selectively affect the target and development of an appropriate method of application become critically important. The scope of this review is to summarize the current concepts and newest advances in research of the immunomodulatory activity of natural and synthetic AMPs, and to examine the prospects and limitations of their medical use.
Collapse
|
8
|
Wollny T, Daniluk T, Piktel E, Wnorowska U, Bukłaha A, Głuszek K, Durnaś B, Bucki R. Targeting the Gut Microbiota to Relieve the Symptoms of Irritable Bowel Syndrome. Pathogens 2021; 10:1545. [PMID: 34959500 PMCID: PMC8705654 DOI: 10.3390/pathogens10121545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a common, chronic, functional disorder with a large impact on world population. Its pathophysiology is not completely revealed; however, it is certain that dysregulation of the bidirectional communications between the central nervous system (CNS) and the gut leads to motility disturbances, visceral hypersensitivity, and altered CNS processing characterized by differences in brain structure, connectivity and functional responsiveness. Emerging evidence suggests that gut microbiota exerts a marked influence on the host during health and disease. Gut microbiome disturbances can be also important for development of IBS symptoms and its modulation efficiently contributes to the therapy. In this work, we review the current knowledge about the IBS therapy, the role of gut microbiota in pathogenesis of IBS, and we discuss that its targeting may have significant impact on the effectiveness of IBS therapy.
Collapse
Affiliation(s)
- Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734 Kielce, Poland;
| | - Tamara Daniluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland; (T.D.); (E.P.); (U.W.)
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland; (T.D.); (E.P.); (U.W.)
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland; (T.D.); (E.P.); (U.W.)
| | - Anna Bukłaha
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Białystok, Waszyngtona 15a, 15-269 Białystok, Poland;
| | - Katarzyna Głuszek
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, 25-734 Kielce, Poland; (K.G.); (B.D.)
| | - Bonita Durnaś
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, 25-734 Kielce, Poland; (K.G.); (B.D.)
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland; (T.D.); (E.P.); (U.W.)
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, 25-734 Kielce, Poland; (K.G.); (B.D.)
| |
Collapse
|
9
|
Zhou C, Zhou X, Wen Z, Liu L, Yang Z, Yang L, Li P, Guo X, Mei X. Compound Fu brick tea modifies the intestinal microbiome composition in high-fat diet-induced obesity mice. Food Sci Nutr 2020; 8:5508-5520. [PMID: 33133553 PMCID: PMC7590332 DOI: 10.1002/fsn3.1850] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/17/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Compound Fu Brick Tea (CFBT), which is from Duyun city in China, is a traditional Chinese dark tea, Fu Brick Tea, mixed with six herbal medicine. It is consumed by local people for reducing weight, but the mechanism is not clear. The disorder of intestinal microbiome caused by long-term high-fat diet (HFD) is one of the inducements of obesity and related metabolic syndrome. In this study, mice were fed with HFD to establish a high-fat model. Fifty mice were randomly divided into six groups: normal control (CK), HFD model control (NK), positive control with medicine (YK), CFBT groups with low, middle, and high dose (FL, FM, FH). The V3-V4 DNA region of fecal microbiome from mouse intestine was sequenced. The results showed that the diversity of intestinal microflora was highest in CK and lowest in NK. Compared with CK, the dominant bacterium Firmicutes was increased and Bacteroidetes decreased at phylum level in NK. Compared with NK, the abundance of microbiome in CFBT groups was significantly higher and the composition was changed: Muribaculaceae, Bacteroidaceae, and Prevotellaceae increased and Lachnospiraceae decreased in CFBT groups at family level, while at the genus level, Bacteroides increased and Lactobacillus decreased. These results conclude that CFBT can increase the abundance of intestinal microbiome in mice, promote the growth of beneficial bacteria and reduce the number of pathogenic bacteria, and restore the imbalance of intestinal microbiome caused by poor diet.
Collapse
Affiliation(s)
- Caibi Zhou
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Xiaolu Zhou
- College of Horticulture and Landscape ArchitectureHunan Agricultural UniversityChangshaChina
| | - Zhirui Wen
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Liming Liu
- College of Horticulture and Landscape ArchitectureHunan Agricultural UniversityChangshaChina
| | - Zaibo Yang
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Lu Yang
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Ping Li
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Xiying Guo
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| | - Xin Mei
- College of Biological Science and AgricultureQiannan Normal University for NationalitiesDuyunChina
| |
Collapse
|
10
|
Panwar H, Rokana N, Aparna SV, Kaur J, Singh A, Singh J, Singh KS, Chaudhary V, Puniya AK. Gastrointestinal stress as innate defence against microbial attack. J Appl Microbiol 2020; 130:1035-1061. [PMID: 32869386 DOI: 10.1111/jam.14836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/09/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022]
Abstract
The human gastrointestinal (GI) tract has been bestowed with the most difficult task of protecting the underlying biological compartments from the resident commensal flora and the potential pathogens in transit through the GI tract. It has a unique environment in which several defence tactics are at play while maintaining homeostasis and health. The GI tract shows myriad number of environmental extremes, which includes pH variations, anaerobic conditions, nutrient limitations, elevated osmolarity etc., which puts a check to colonization and growth of nonfriendly microbial strains. The GI tract acts as a highly selective barrier/platform for ingested food and is the primary playground for balance between the resident and uninvited organisms. This review focuses on antimicrobial defense mechanisms of different sections of human GI tract. In addition, the protective mechanisms used by microbes to combat the human GI defence systems are also discussed. The ability to survive this innate defence mechanism determines the capability of probiotic or pathogen strains to confer health benefits or induce clinical events respectively.
Collapse
Affiliation(s)
- H Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - N Rokana
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - S V Aparna
- Department of Dairy Microbiology, College of Dairy Science and Technology, Kerala Veterinary and Animal Science University, Mannuthy, Thrissur, India
| | - J Kaur
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - A Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - J Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - K S Singh
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - V Chaudhary
- Department of Microbiology, Punjab Agriculture University, Ludhiana, Punjab, India
| | - A K Puniya
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
11
|
Siddiqui R, Akbar N, Khan NA. Gut microbiome and human health under the space environment. J Appl Microbiol 2020; 130:14-24. [PMID: 32692438 DOI: 10.1111/jam.14789] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
The gut microbiome is well recognized to have a pivotal role in regulation of the health and behaviour of the host, affecting digestion, metabolism, immunity, and has been linked to changes in bones, muscles and the brain, to name a few. However, the impact of microgravity environment on gut bacteria is not well understood. In space environments, astronauts face several health issues including stress, high iron diet, radiation and being in a closed system during extended space missions. Herein, we discuss the role of gut bacteria in the space environment, in relation to factors such as microgravity, radiation and diet. Gut bacteria may exact their effects by synthesis of molecules, their absorption, and through physiological effects on the host. Moreover we deliberate the role of these challenges in the dysbiosis of the human microbiota and possible dysregulation of the immune system.
Collapse
Affiliation(s)
- R Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| | - N Akbar
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| | - N A Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| |
Collapse
|
12
|
Effect of Antihypertensive Medications on Sepsis-Related Outcomes: A Population-Based Cohort Study. Crit Care Med 2020; 47:e386-e393. [PMID: 30688717 DOI: 10.1097/ccm.0000000000003654] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Although the effect of antihypertensive agents on sepsis has been studied, evidence for survival benefit was limited in the literature. We investigated differences in sepsis-related outcomes depending on the antihypertensive drugs given prior to sepsis in patients with hypertension. DESIGN Population-based cohort study. SETTING Sample cohort Database of the National Health Insurance Service from 2003 to 2013 in South Korea. PATIENTS Patients over 30 years old who were diagnosed with sepsis after receiving hypertension treatment. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Primary outcomes, 30-day and 90-day mortality rates, were analyzed for differences among three representative antihypertensive medications: angiotensin- converting enzyme inhibitors or angiotensin II receptor blockers, calcium channel blockers, and thiazides. In total, 4,549 patients diagnosed with hypertension prior to hospitalization for sepsis were identified. The 30-day mortality was significantly higher among patients who did not receive any medications within 1 month before sepsis (36.8%) than among patients who did (32.0%; p < 0.001). The risk for 90-days mortality was significantly lower in prior angiotensin-converting enzyme inhibitors or angiotensin II receptor blocker users (reference) than in other drug users (odds ratio, 1.27; 95% CI, 1.07-1.52). There was no difference in the risk for 30-day and 90-day mortality depending on whether calcium channel blockers or thiazides were used. Use of calcium channel blockers was associated with a decreased risk for inotropic agent administration, compared with those of angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers (odds ratio, 1.23; 95% CI, 1.05-1.44) and thiazides (odds ratio, 1.33; 95% CI, 1.12-1.58). CONCLUSIONS In patients with sepsis, lower mortality rate was associated with prior use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers not with use of calcium channel blockers or thiazides. The requirement of inotropic agents was significantly lower in prior use of calcium channel blockers, although the survival benefits were not prominent.
Collapse
|
13
|
Balan P, Staincliffe M, Moughan PJ. Dietary supplementation with ovine serum immunoglobulin modulates correlations between mucin, microbiota and immunity proteins in the growing rat. J Anim Physiol Anim Nutr (Berl) 2020; 104:758-766. [DOI: 10.1111/jpn.13319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/10/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Prabhu Balan
- Riddet Institute Massey University Palmerston North New Zealand
- Alpha Massey Natural Nutraceutical Research Centre Palmerston North New Zealand
| | | | - Paul J. Moughan
- Riddet Institute Massey University Palmerston North New Zealand
- Alpha Massey Natural Nutraceutical Research Centre Palmerston North New Zealand
| |
Collapse
|
14
|
Elgin TG, Fricke EM, Gong H, Reese J, Mills DA, Kalantera KM, Underwood MA, McElroy SJ. Fetal exposure to maternal inflammation interrupts murine intestinal development and increases susceptibility to neonatal intestinal injury. Dis Model Mech 2019; 12:dmm.040808. [PMID: 31537532 PMCID: PMC6826024 DOI: 10.1242/dmm.040808] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Fetal exposure to chorioamnionitis can impact the outcomes of the developing fetus both at the time of birth and in the subsequent neonatal period. Infants exposed to chorioamnionitis have a higher incidence of gastrointestinal (GI) pathology, including necrotizing enterocolitis (NEC); however, the mechanism remains undefined. To simulate the fetal exposure to maternal inflammation (FEMI) induced by chorioamnionitis, pregnant mice (C57BL/6J, IL-6 -/-, RAG -/- or TNFR1 -/-) were injected intraperitoneally on embryonic day (E)15.5 with lipopolysaccharide (LPS; 100 µg/kg body weight). Pups were delivered at term, and reared to postnatal day (P)0, P7, P14, P28 or P56. Serum and intestinal tissue samples were collected to quantify growth, inflammatory markers, histological intestinal injury, and goblet and Paneth cells. To determine whether FEMI increased subsequent susceptibility to intestinal injury, a secondary dose of LPS (100 µg/kg body weight) was given on P5, prior to tissue harvesting on P7. FEMI had no effect on growth of the offspring or their small intestine. FEMI significantly decreased both goblet and Paneth cell numbers while simultaneously increasing serum levels of IL-1β, IL-10, KC/GRO (CXCL1 and CXCL2), TNF and IL-6. These alterations were IL-6 dependent and, importantly, increased susceptibility to LPS-induced intestinal injury later in life. Our data show that FEMI impairs normal intestinal development by decreasing components of innate immunity and simultaneously increasing markers of inflammation. These changes increase susceptibility to intestinal injury later in life and provide novel mechanistic data to potentially explain why preterm infants exposed to chorioamnionitis prior to birth have a higher incidence of NEC and other GI disorders.
Collapse
Affiliation(s)
- Timothy G Elgin
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Erin M Fricke
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Huiyu Gong
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Jeffrey Reese
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232, USA
| | - David A Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA
| | - Karen M Kalantera
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA
| | - Mark A Underwood
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA
| | - Steven J McElroy
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA .,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
15
|
Yang L, Liu G, Lian K, Qiao Y, Zhang B, Zhu X, Luo Y, Shang Y, Gu XL. Dietary leonurine hydrochloride supplementation attenuates lipopolysaccharide challenge-induced intestinal inflammation and barrier dysfunction by inhibiting the NF-κB/MAPK signaling pathway in broilers. J Anim Sci 2019; 97:1679-1692. [PMID: 30789669 DOI: 10.1093/jas/skz078] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/20/2019] [Indexed: 12/20/2022] Open
Abstract
This study was performed to evaluate the beneficial effects of dietary leonurine hydrochloride (LH) supplementation on intestinal morphology and barrier integrity and further illuminate its underlying antioxidant and immunomodulatory mechanisms in lipopolysaccharide (LPS)-treated broilers. A total of 120 1-d-old male broilers (Ross 308) were assigned to 4 treatment groups with 6 replicates of 5 birds per cage. The experiment was designed in a 2 × 2 factorial arrangement with LH (0 or 120 mg/kg) and LPS (injection of saline or 1.5 mg/kg body weight) as treatments. On days 14, 16, 18, and 20 of the trial, broilers were intraperitoneally injected with LPS or physiological saline. Compared with the control group, LPS-challenged broilers showed impaired growth performance (P < 0.05) from day 15 to day 21 of the trial, increased serum diamine oxidase (DAO) and D-lactic acid (D-LA) levels coupled with reduced glutathione (GSH) content and total superoxide dismutase (T-SOD) activity (duodenal and jejunal mucosa), reduced malondialdehyde (MDA) content (duodenal, jejunal, and ileal mucosa), and compromised morphological structure of the duodenum and jejunum. Additionally, LPS challenge increased (P < 0.05) the mRNA expression of proinflammatory cytokine genes and reduced tight junction (TJ) protein expression in the jejunum. However, dietary LH prevented LPS-induced reductions in average daily gain (ADG) and average daily feed intake (ADFI) in broilers. It also alleviated LPS challenge-induced increases in serum DAO levels, MDA content (duodenal and jejunal mucosa), and jejunal crypt depth (P < 0.05) but reduced villus height, GSH content (jejunal mucosa), and T-SOD activity (duodenal and jejunal mucosa) (P < 0.05). Additionally, LH supplementation significantly downregulated the mRNA expression of nuclear factor (NF)-κB, cyclooxygenase-2 (COX-2), and proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and upregulated the mRNA expression of zonula occludens-1 (ZO-1) and Occludin in the jejunal mucosa induced by LPS (P < 0.05). On the other hand, LH administration prevented LPS-induced activation of the p38, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) and attenuated IkB alpha (IκBα) phosphorylation and nuclear translocation of NF-κB (p65) in the jejunal mucosa. In conclusion, dietary LH supplementation attenuates intestinal mucosal disruption mainly by accelerating the expression of TJ proteins and inhibiting activation of the NF-κB/MAPK signaling pathway.
Collapse
Affiliation(s)
- Li Yang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Gang Liu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Kexun Lian
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yanjie Qiao
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Baojun Zhang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Xiaoqing Zhu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yan Luo
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yunxia Shang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Xin-Li Gu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| |
Collapse
|
16
|
Gunasekara DB, DiSalvo M, Wang Y, Nguyen DL, Reed MI, Speer J, Sims CE, Magness ST, Allbritton NL. Development of Arrayed Colonic Organoids for Screening of Secretagogues Associated with Enterotoxins. Anal Chem 2018; 90:1941-1950. [PMID: 29281259 PMCID: PMC6028038 DOI: 10.1021/acs.analchem.7b04032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Enterotoxins increase intestinal fluid secretion through modulation of ion channels as well as activation of the enteric nervous and immune systems. Colonic organoids, also known as colonoids, are functionally and phenotypically similar to in vivo colonic epithelium and have been used to study intestinal ion transport and subsequent water flux in physiology and disease models. In conventional cultures, organoids exist as spheroids embedded within a hydrogel patty of extracellular matrix, and they form at multiple depths, impairing efficient imaging necessary to capture data from statistically relevant sample sizes. To overcome these limitations, an analytical platform with colonic organoids localized to the planar surface of a hydrogel layer was developed. The arrays of densely packed colonoids (140 μm average diameter, 4 colonoids/mm2) were generated in a 96-well plate, enabling assay of the response of hundreds of organoids so that organoid subpopulations with distinct behaviors were identifiable. Organoid cell types, monolayer polarity, and growth were similar to those embedded in hydrogel. An automated imaging and analysis platform efficiently tracked over time swelling due to forskolin and fluid movement across the cell monolayer stimulated by cholera toxin. The platform was used to screen compounds associated with the enteric nervous and immune systems for their effect on fluid movement across epithelial cells. Prostaglandin E2 promoted increased water flux in a subset of organoids that resulted in organoid swelling, confirming a role for this inflammatory mediator in diarrheal conditions but also illustrating organoid differences in response to an identical stimulus. By allowing sampling of a large number of organoids, the arrayed organoid platform permits identification of organoid subpopulations intermixed within a larger group of nonresponding organoids. This technique will enable automated, large-scale screening of the impact of drugs, toxins, and other compounds on colonic physiology.
Collapse
Affiliation(s)
- Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Daniel L. Nguyen
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Mark I. Reed
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
17
|
Lim H, Cun W, Wang Y, Gray RA, Glimm J. The role of conductivity discontinuities in design of cardiac defibrillation. CHAOS (WOODBURY, N.Y.) 2018; 28:013106. [PMID: 29390616 DOI: 10.1063/1.5019367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Fibrillation is an erratic electrical state of the heart, of rapid twitching rather than organized contractions. Ventricular fibrillation is fatal if not treated promptly. The standard treatment, defibrillation, is a strong electrical shock to reinitialize the electrical dynamics and allow a normal heart beat. Both the normal and the fibrillatory electrical dynamics of the heart are organized into moving wave fronts of changing electrical signals, especially in the transmembrane voltage, which is the potential difference between the cardiac cellular interior and the intracellular region of the heart. In a normal heart beat, the wave front motion is from bottom to top and is accompanied by the release of Ca ions to induce contractions and pump the blood. In a fibrillatory state, these wave fronts are organized into rotating scroll waves, with a centerline known as a filament. Treatment requires altering the electrical state of the heart through an externally applied electrical shock, in a manner that precludes the existence of the filaments and scroll waves. Detailed mechanisms for the success of this treatment are partially understood, and involve local shock-induced changes in the transmembrane potential, known as virtual electrode alterations. These transmembrane alterations are located at boundaries of the cardiac tissue, including blood vessels and the heart chamber wall, where discontinuities in electrical conductivity occur. The primary focus of this paper is the defibrillation shock and the subsequent electrical phenomena it induces. Six partially overlapping causal factors for defibrillation success are identified from the literature. We present evidence in favor of five of these and against one of them. A major conclusion is that a dynamically growing wave front starting at the heart surface appears to play a primary role during defibrillation by critically reducing the volume available to sustain the dynamic motion of scroll waves; in contrast, virtual electrodes occurring at the boundaries of small, isolated blood vessels only cause minor effects. As a consequence, we suggest that the size of the heart (specifically, the surface to volume ratio) is an important defibrillation variable.
Collapse
Affiliation(s)
- Hyunkyung Lim
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York 11794-3600, USA
| | - Wenjing Cun
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York 11794-3600, USA
| | - Yue Wang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York 11794-3600, USA
| | - Richard A Gray
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland 20993-0002, USA
| | - James Glimm
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York 11794-3600, USA
| |
Collapse
|
18
|
Jin L, Wang W, Degroote J, Van Noten N, Yan H, Majdeddin M, Van Poucke M, Peelman L, Goderis A, Van De Mierop K, Mombaerts R, De Smet S, Michiels J. Mycotoxin binder improves growth rate in piglets associated with reduction of toll-like receptor-4 and increase of tight junction protein gene expression in gut mucosa. J Anim Sci Biotechnol 2017; 8:80. [PMID: 29118977 PMCID: PMC5664444 DOI: 10.1186/s40104-017-0210-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/04/2017] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Deoxynivalenol (DON) is a mycotoxin produced by Fusarium species in the field, commonly found in cereal grains, which negatively affects performances and health of animals. Mycotoxin binders are supposed to reduce the toxicity of mycotoxins. METHOD The effect of a mycotoxin binder (containing acid-activated bentonite, clinoptilolite, yeast cell walls and organic acids) on growth performance and gut health was studied. Hundred and twenty weaning piglets were allocated to 4 treatments, with 5 pens of 6 piglets each, arranged in a 2 × 2 factorial design: control diet; control diet with 1 kg/t binder; control diet with DON; and control diet with DON and 1 kg/t binder. From d0-14, the diet of DON-challenged groups was artificially contaminated with a mixture of DON (2.6 mg/kg), 3-acetyl-deoxynivalenol (0.1 mg/kg) and 15-acetyl-deoxynivalenol (0.3 mg/kg), after which the total contamination level was reduced to 1 mg/kg, until d37. On d14, one pig from each pen was euthanized and distal small intestinal mucosa samples were collected for the assessment of intestinal permeability, and gene expression of tight junction proteins, toll-like receptor 4, inflammatory cytokines and intestinal alkaline phosphatase. RESULTS After 37 d, there were no differences in growth performance between control and DON-challenged groups (P > 0.05). Nevertheless, groups that received diets with binder had a significantly higher average daily gain (ADG) and average daily feed intake (ADFI) for the first 14 d as well as for the whole period, compared to groups without binder (P ≤ 0.05). Groups with binder in the diet also exhibited lower expression of toll-like receptor 4 in distal small intestinal mucosa at d14, compared to groups without binder (P ≤ 0.05). Interestingly, comparing the two DON treatments, piglets fed DON and binder had significantly higher ADFI and ADG compared to those with only DON for the first 14-d (P ≤ 0.05). Addition of binder to DON contaminated diets, also down-regulated the gene expression of toll-like receptor 4 (P ≤ 0.05) and increased mRNA level zona occludens 1 (P ≤ 0.10) as compared to DON. CONCLUSIONS The present data provide evidence that the binder improves growth rate in piglets associated with reduction of toll-like receptor-4 and increase of tight junction protein gene expression. However, the current study does not allow to assess whether the effects of the binder are mediated by alterations in the toxicokinetics of the mycotoxin.
Collapse
Affiliation(s)
- Linghong Jin
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Wei Wang
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Jeroen Degroote
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Noémie Van Noten
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Honglin Yan
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Maryam Majdeddin
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Mario Van Poucke
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium
| | | | | | | | - Stefaan De Smet
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Production, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Joris Michiels
- Department of Applied Biosciences, Ghent University, Valentin Vaerwyckweg 1, 9000 Ghent, Belgium
| |
Collapse
|
19
|
Preadmission Use of Calcium Channel Blocking Agents Is Associated With Improved Outcomes in Patients With Sepsis: A Population-Based Propensity Score-Matched Cohort Study. Crit Care Med 2017; 45:1500-1508. [PMID: 28658023 DOI: 10.1097/ccm.0000000000002550] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Use of calcium channel blockers has been found to improve sepsis outcomes in animal studies and one clinical study. This study determines whether the use of calcium channel blockers is associated with a decreased risk of mortality in patients with sepsis. DESIGN Population-based matched cohort study. SETTING National Health Insurance Research Database of Taiwan. PATIENTS Hospitalized severe sepsis patients identified from National Health Insurance Research Database by International Classification of Diseases, Ninth Revision, Clinical Modification codes. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The association between calcium channel blocker use and sepsis outcome was determined by multivariate-adjusted Cox proportional hazard models and propensity score analysis. To examine the influence of healthy user bias, beta-blocker was used as an active comparator. Our study identified 51,078 patients with sepsis, of which, 19,742 received calcium channel blocker treatments prior to the admission. Use of calcium channel blocker was associated with a reduced 30-day mortality after propensity score adjustment (hazard ratio, 0.94; 95% CI, 0.89-0.99), and the beneficial effect could extend to 90-day mortality (hazard ratio, 0.95; 95% CI, 0.89-1.00). In contrast, use of beta-blocker was not associated with an improved 30-day (hazard ratio, 1.06; 95% CI, 0.97-1.15) or 90-day mortality (hazard ratio, 1.00; 95% CI, 0.90-1.11). On subgroup analysis, calcium channel blockers tend to be more beneficial to patients with male gender, between 40 and 79 years old, with a low comorbidity burden, and to patients with cardiovascular diseases, diabetes, or renal diseases. CONCLUSIONS In this national cohort study, preadmission calcium channel blocker therapy before sepsis development was associated with a 6% reduction in mortality when compared with patients who have never received calcium channel blockers.
Collapse
|
20
|
Wang W, Li Z, Lv Z, Zhang B, Lv H, Guo Y. Effects of Kluyveromyces marxianus supplementation on immune responses, intestinal structure and microbiota in broiler chickens. PLoS One 2017; 12:e0180884. [PMID: 28700666 PMCID: PMC5507273 DOI: 10.1371/journal.pone.0180884] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/22/2017] [Indexed: 01/31/2023] Open
Abstract
To investigate the effects of Kluyveromyces marxianus on immune responses, intestinal structure and microbiota in broilers, 840 1-d-old broiler chicks were randomly divided into seven groups (eight replicates) and were fed basal diets without or with 0.25, 0.50, 1.0, 1.5, 2.0, and 2.5 g/kg of K. marxianus (2.0×1010 CFU/g). Serum and intestine samples were collected at 21 d of age. The results showed that increasing K. marxianus addition linearly reduced feed conversion ratio but linearly elevated relative thymus weight, as well as quadratically increased serum lysozyme and IgG levels, with the medium dose (1.0 g/kg) being the most effective. The ratio of villus height to crypt depth of jejunum and ileum, ileal villus height and sucrase activity, as well as the mRNA expression of ileal mucin-2, claudin-1 and sodium glucose cotransporter 1 linearly responded to the increasing K. marxianus addition. Supplemental K. marxianus at low (0.5 g/kg), medium (1.5 g/kg) and high (2.5 g/kg) dose all decreased the abundance of phylum Cyanobacteria, increased the abundance of phylum Firmicutes and genus Lactobacillus in ileum. The high dose of K. marxianus addition also reduced the abundance of order Rickettsiales and Pseudomonadales along with species Acinetobacter junii. Ileal bacterial communities between K. marxianus-treated and untreated groups formed distinctly different clusters. In summary, K. marxianus supplementation benefits feed efficiency and immune function, as well as intestinal structure in broilers, which might be attributed to the improved ileal microbial structure. Supplemental K. marxianus at high dose (2.5 g/kg) was more effective for feed efficiency and intestinal health of broilers, while the innate immunity was optimized at a medium dose (1.0 g/kg).
Collapse
Affiliation(s)
- Weiwei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhui Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Beibei Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Hong Lv
- Shanghai Engineering Research Center of Industrial Microorganisms, College of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
21
|
Abreu MT, Thomas LS, Arnold ET, Lukasek K, Michelsen KS, Arditi M. TLR signaling at the intestinal epithelial interface. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519030090050901] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The intestinal epithelium provides a critical interface between lumenal bacteria and the mucosal immune system. Whereas normal commensal flora do not trigger acute inflammation, pathogenic bacteria trigger a potent inflammatory response. Our studies emanate from the hypothesis that the intestinal epithelium is normally hyporesponsive to commensal pathogen-associated molecular patterns (PAMPs) such as LPS. Our data demonstrate that normal human colonic epithelial cells and lamina propria cells express low levels of TLR4 and its co-receptor MD-2. This expression pattern is mirrored by intestinal epithelial cell (IEC) lines. Co-expression of TLR4 and MD-2 is necessary and sufficient for LPS responsiveness in IEC. Moreover, LPS sensing occurs along the basolateral membrane of polarized IEC in culture. Expression of MD-2 is regulated by IFN-γ. Cloning of the MD-2 promoter demonstrates that promoter activity is increased by IFN-γ and blocked by the STAT inhibitor SOCS3. We conclude from our studies that the intestinal epithelium down-regulates expression of TLR4 and MD-2 and is LPS unresponsive. The Th1 cytokine IFN-γ up-regulates expression of MD-2 in a STAT-dependent fashion. The results of our studies have important implications for understanding human inflammatory bowel diseases.
Collapse
Affiliation(s)
- Maria T. Abreu
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA,
| | - Lisa S. Thomas
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elizabeth T. Arnold
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Katie Lukasek
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kathrin S. Michelsen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Steven Spielberg Pediatric Research Center, Burns and Allen Research Institute
| | - Moshe Arditi
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Steven Spielberg Pediatric Research Center, Burns and Allen Research Institute
| |
Collapse
|
22
|
Han F, Lu Z, Liu Y, Xia X, Zhang H, Wang X, Wang Y. Cathelicidin-BF ameliorates lipopolysaccharide-induced intestinal epithelial barrier disruption in rat. Life Sci 2016; 152:199-209. [DOI: 10.1016/j.lfs.2016.03.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/21/2016] [Accepted: 03/21/2016] [Indexed: 12/21/2022]
|
23
|
O'Shea CJ, O'Doherty JV, Callanan JJ, Doyle D, Thornton K, Sweeney T. The effect of algal polysaccharides laminarin and fucoidan on colonic pathology, cytokine gene expression and Enterobacteriaceae in a dextran sodium sulfate-challenged porcine model. J Nutr Sci 2016; 5:e15. [PMID: 27110358 PMCID: PMC4831127 DOI: 10.1017/jns.2016.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 12/14/2015] [Accepted: 01/11/2016] [Indexed: 01/02/2023] Open
Abstract
The algal polysaccharides laminarin (LAM) and fucoidan (FUC) have potent anti-inflammatory activities in the gastrointestinal tract. Our objective was to examine the impact of prior consumption of LAM and/or FUC on pathology and inflammation following a dextran sodium sulfate (DSS) challenge in pigs. Pigs (n 7/group) were assigned to one of five experimental groups for 56 d. From 49-55 d, distilled water or DSS was administered intragastrically. The experimental groups were: (1) basal diet + distilled water (control); (2) basal diet + DSS (DSS); (3) basal diet + FUC + DSS (FUC + DSS); (4) basal diet + LAM + DSS (LAM + DSS); and (5) basal diet + LAM + FUC + DSS (LAMFUC + DSS). The DSS group had decreased body-weight gain (P < 0·05) and serum xylose (P < 0·05), and increased proximal colon pathology score (P < 0·05), diarrhoeal score (P < 0·001) and colonic Enterobacteriaceae (P < 0·05) relative to the control group. The FUC + DSS (P < 0·01), LAM + DSS (P < 0·05) and LAMFUC + DSS (P < 0·05) groups had improved diarrhoeal score, and the LAMFUC + DSS (P < 0·05) group had improved body weight relative to the DSS group. The FUC + DSS group (P < 0·001), LAM + DSS group (P < 0·05) and LAMFUC + DSS group (P < 0·001) had lower IL-6 mRNA abundance relative to the DSS group. The LAM + DSS group had reduced Enterobacteriaceae in proximal colon digesta relative to the DSS group (P < 0·05). In conclusion, FUC or a combination of FUC and LAM improved body-weight loss, diarrhoeal scores and clinical variables associated with a DSS challenge in pigs, in tandem with a reduction in colonic IL-6 mRNA abundance.
Collapse
Affiliation(s)
- C. J. O'Shea
- School of Agriculture and Food Science, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - J. V. O'Doherty
- School of Agriculture and Food Science, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - J. J. Callanan
- School of Veterinary Medicine, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - D. Doyle
- School of Agriculture and Food Science, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - K. Thornton
- School of Veterinary Medicine, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - T. Sweeney
- School of Veterinary Medicine, College of Life Sciences, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| |
Collapse
|
24
|
Fu W, Chen L, Wang Z, Zhao C, Chen G, Liu X, Dai Y, Cai Y, Li C, Zhou J, Liang G. Determination of the binding mode for anti-inflammatory natural product xanthohumol with myeloid differentiation protein 2. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:455-63. [PMID: 26869767 PMCID: PMC4737557 DOI: 10.2147/dddt.s98466] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is recognized that myeloid differentiation protein 2 (MD-2), a coreceptor of toll-like receptor 4 (TLR4) for innate immunity, plays an essential role in activation of the lipopolysaccharide signaling pathway. MD-2 is known as a neoteric and suitable therapeutical target. Therefore, there is great interest in the development of a potent MD-2 inhibitor for anti-inflammatory therapeutics. Several studies have reported that xanthohumol (XN), an anti-inflammatory natural product from hops and beer, can block the TLR4 signaling by binding to MD-2 directly. However, the interaction between MD-2 and XN remains unknown. Herein, our work aims at characterizing interactions between MD-2 and XN. Using a combination of experimental and theoretical modeling analysis, we found that XN can embed into the hydrophobic pocket of MD-2 and form two stable hydrogen bonds with residues ARG-90 and TYR-102 of MD-2. Moreover, we confirmed that ARG-90 and TYR-102 were two necessary residues during the recognition process of XN binding to MD-2. Results from this study identified the atomic interactions between the MD-2 and XN, which will contribute to future structural design of novel MD-2-targeting molecules for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Weitao Fu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Lingfeng Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Zhe Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Chengwei Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Xing Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Yuanrong Dai
- Department of Respiratory Medicine, the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yuepiao Cai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Chenglong Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China; Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Jianmin Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
25
|
Catecholamine-Directed Epithelial Cell Interactions with Bacteria in the Intestinal Mucosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:79-99. [DOI: 10.1007/978-3-319-20215-0_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
26
|
MacEachern SJ, Patel BA, Keenan CM, Dicay M, Chapman K, McCafferty DM, Savidge TC, Beck PL, MacNaughton WK, Sharkey KA. Inhibiting Inducible Nitric Oxide Synthase in Enteric Glia Restores Electrogenic Ion Transport in Mice With Colitis. Gastroenterology 2015; 149:445-55.e3. [PMID: 25865048 PMCID: PMC4516675 DOI: 10.1053/j.gastro.2015.04.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/06/2015] [Accepted: 04/06/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Disturbances in the control of ion transport lead to epithelial barrier dysfunction in patients with colitis. Enteric glia regulate intestinal barrier function and colonic ion transport. However, it is not clear whether enteric glia are involved in epithelial hyporesponsiveness. We investigated enteric glial regulation of ion transport in mice with trinitrobenzene sulfonic acid- or dextran sodium sulfate-induced colitis and in Il10(-/-) mice. METHODS Electrically evoked ion transport was measured in full-thickness segments of colon from CD1 and Il10(-/-) mice with or without colitis in Ussing chambers. Nitric oxide (NO) production was assessed using amperometry. Bacterial translocation was investigated in the liver, spleen, and blood of mice. RESULTS Electrical stimulation of the colon evoked a tetrodotoxin-sensitive chloride secretion. In mice with colitis, ion transport almost completely disappeared. Inhibiting inducible NO synthase (NOS2), but not neuronal NOS (NOS1), partially restored the evoked secretory response. Blocking glial function with fluoroacetate, which is not a NOS2 inhibitor, also partially restored ion transport. Combined NOS2 inhibition and fluoroacetate administration fully restored secretion. Epithelial responsiveness to vasoactive intestinal peptide was increased after enteric glial function was blocked in mice with colitis. In colons of mice without colitis, NO was produced in the myenteric plexus almost completely via NOS1. NO production was increased in mice with colitis, compared with mice without colitis; a substantial proportion of NOS2 was blocked by fluoroacetate administration. Inhibition of enteric glial function in vivo reduced the severity of trinitrobenzene sulfonic acid-induced colitis and associated bacterial translocation. CONCLUSIONS Increased production of NOS2 in enteric glia contributes to the dysregulation of intestinal ion transport in mice with colitis. Blocking enteric glial function in these mice restores epithelial barrier function and reduces bacterial translocation.
Collapse
Affiliation(s)
- Sarah J. MacEachern
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bhavik A. Patel
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Brighton, UK
| | - Catherine M. Keenan
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael Dicay
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kevin Chapman
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Donna-Marie McCafferty
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tor C. Savidge
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Paul L. Beck
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K. MacNaughton
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A. Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
27
|
Chaiyadet S, Smout M, Johnson M, Whitchurch C, Turnbull L, Kaewkes S, Sotillo J, Loukas A, Sripa B. Excretory/secretory products of the carcinogenic liver fluke are endocytosed by human cholangiocytes and drive cell proliferation and IL6 production. Int J Parasitol 2015; 45:773-81. [PMID: 26187786 DOI: 10.1016/j.ijpara.2015.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/18/2015] [Accepted: 06/06/2015] [Indexed: 01/22/2023]
Abstract
Liver fluke infection caused by Opisthorchis viverrini remains a major public health problem in many parts of Asia including Thailand, Lao PDR, Vietnam and Cambodia, where there is a strikingly high incidence of cholangiocarcinoma (CCA - hepatic cancer of the bile duct epithelium). Among other factors, uptake of O. viverrini excretory/secretory products (OvES) by biliary epithelial cells has been postulated to be responsible for chronic inflammation and proliferation of cholangiocytes, but the mechanisms by which cells internalise O. viverrini excretory/secretory products are still unknown. Herein we incubated normal human cholangiocytes (H69), human cholangiocarcinoma cells (KKU-100, KKU-M156) and human colon cancer (Caco-2) cells with O. viverrini excretory/secretory products and analysed the effects of different endocytic inhibitors to address the mechanism of cellular uptake of ES proteins. Opisthorchis viverrini excretory/secretory products was internalised preferentially by liver cell lines, and most efficiently/rapidly by H69 cells. There was no evidence for trafficking of ES proteins to cholangiocyte organelles, and most of the fluorescence was detected in the cytoplasm. Pretreatment with clathrin inhibitors significantly reduced the uptake of O. viverrini excretory/secretory products, particularly by H69 cells. Opisthorchis viverrini excretory/secretory products induced proliferation of liver cells (H69 and CCA lines) but not intestinal (Caco-2) cells, and proliferation was blocked using inhibitors of the classical endocytic pathways (clathrin and caveolae). Opisthorchis viverrini excretory/secretory products drove IL6 secretion by H69 cells but not Caco-2 cells, and cytokine secretion was significantly reduced by endocytosis inhibitors. This the first known study to address the endocytosis of helminth ES proteins by host epithelial cells and sheds light on the pathways by which this parasite causes one of the most devastating forms of cancer in south-eastern Asia.
Collapse
Affiliation(s)
- Sujittra Chaiyadet
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand; Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Michael Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Michael Johnson
- Faculty of Science, University of Technology Sydney, Sydney, Australia
| | | | - Lynne Turnbull
- Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Sasithorn Kaewkes
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Banchob Sripa
- Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
28
|
|
29
|
The role of toll-like receptors 2 and 4 in the pathogenesis of feline pyometra. Theriogenology 2015; 83:596-603. [DOI: 10.1016/j.theriogenology.2014.10.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/17/2014] [Accepted: 10/27/2014] [Indexed: 11/21/2022]
|
30
|
Halpern MD, Denning PW. The role of intestinal epithelial barrier function in the development of NEC. Tissue Barriers 2015; 3:e1000707. [PMID: 25927016 DOI: 10.1080/21688370.2014.1000707] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/16/2014] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelial barrier plays an important role in maintaining host health. Breakdown of intestinal barrier function is known to play a role in many diseases such as infectious enteritis, idiopathic inflammatory bowel disease, and neonatal inflammatory bowel diseases. Recently, increasing research has demonstrated the importance of understanding how intestinal epithelial barrier function develops in the premature neonate in order to develop strategies to promote its maturation. Optimizing intestinal barrier function is thought to be key to preventing neonatal inflammatory bowel diseases such as necrotizing enterocolitis. In this review, we will first summarize the key components of the intestinal epithelial barrier, what is known about its development, and how this may explain NEC pathogenesis. Finally, we will review what therapeutic strategies may be used to promote optimal development of neonatal intestinal barrier function in order to reduce the incidence and severity of NEC.
Collapse
Key Words
- AJ, adherens junctions
- AJC, apical junction complex
- BAs, bile acids; EGF, epidermal growth factor
- Bb, Bifidobacterium bifidum
- Bi, Bifidobacterium infantis
- EPO, erythropoietin; IFNγ, interferon gamma
- IEL, intestinal epithelial lymphocytes
- NEC, necrotizing enterocolitis
- NICU, neonatal intensive care unit
- Necrotizing enterocolitis (NEC)
- TCRγδ, T-cell receptor gamma-delta
- TGF-β, transforming growth factor-beta
- TJ, tight junctions
- TNFα, tumor necrosis factor alpha
- TPN, total parenteral nutrition
- apical junctional complex
- commensal bacteria
- intestinal epithelial barrier
- prematurity
- probiotics
- tight junctions
Collapse
Affiliation(s)
- Melissa D Halpern
- Department of Pediatrics; The University of Arizona ; Tucson, AZ USA
| | - Patricia W Denning
- Department of Pediatrics; Emory University School of Medicine ; Atlanta, GA USA
| |
Collapse
|
31
|
Imaging of Ventricular Fibrillation and Defibrillation: The Virtual Electrode Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 859:343-65. [PMID: 26238060 DOI: 10.1007/978-3-319-17641-3_14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Ventricular fibrillation is the major underlying cause of sudden cardiac death. Understanding the complex activation patterns that give rise to ventricular fibrillation requires high resolution mapping of localized activation. The use of multi-electrode mapping unraveled re-entrant activation patterns that underlie ventricular fibrillation. However, optical mapping contributed critically to understanding the mechanism of defibrillation, where multi-electrode recordings could not measure activation patterns during and immediately after a shock. In addition, optical mapping visualizes the virtual electrodes that are generated during stimulation and defibrillation pulses, which contributed to the formulation of the virtual electrode hypothesis. The generation of virtual electrode induced phase singularities during defibrillation is arrhythmogenic and may lead to the induction of fibrillation subsequent to defibrillation. Defibrillating with low energy may circumvent this problem. Therefore, the current challenge is to use the knowledge provided by optical mapping to develop a low energy approach of defibrillation, which may lead to more successful defibrillation.
Collapse
|
32
|
Graves CL, Harden SW, LaPato M, Nelson M, Amador B, Sorenson H, Frazier CJ, Wallet SM. A method for high purity intestinal epithelial cell culture from adult human and murine tissues for the investigation of innate immune function. J Immunol Methods 2014; 414:20-31. [PMID: 25193428 PMCID: PMC4384334 DOI: 10.1016/j.jim.2014.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 12/29/2022]
Abstract
Intestinal epithelial cells (IECs) serve as an important physiologic barrier between environmental antigens and the host intestinal immune system. Thus, IECs serve as a first line of defense and may act as sentinel cells during inflammatory insults. Despite recent renewed interest in IEC contributions to host immune function, the study of primary IEC has been hindered by lack of a robust culture technique, particularly for small intestinal and adult tissues. Here, a novel adaptation for culture of primary IEC is described for human duodenal organ donor tissue as well as duodenum and colon of adult mice. These epithelial cell cultures display characteristic phenotypes and are of high purity. In addition, the innate immune function of human primary IEC, specifically with regard to Toll-like receptor (TLR) expression and microbial ligand responsiveness, is contrasted with a commonly used intestinal epithelial cell line (HT-29). Specifically, TLR expression at the mRNA level and production of cytokine (IFNγ and TNFα) in response to TLR agonist stimulation is assessed. Differential expression of TLRs as well as innate immune responses to ligand stimulation is observed in human-derived cultures compared to that of HT-29. Thus, use of this adapted method to culture primary epithelial cells from adult human donors and from adult mice will allow for more appropriate studies of IECs as innate immune effectors.
Collapse
Affiliation(s)
- Christina L Graves
- Department of Oral Biology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL, USA; Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| | - Scott W Harden
- Department of Oral Biology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, P.O. Box 100244, Gainesville 32610, FL, USA.
| | - Melissa LaPato
- Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| | - Michael Nelson
- Department of Oral Biology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL, USA; Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| | - Byron Amador
- Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| | - Heather Sorenson
- Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| | - Charles J Frazier
- Department of Neuroscience, College of Medicine, University of Florida, P.O. Box 100244, Gainesville 32610, FL, USA; Department of Pharmacodynamics, College of Medicine, University of Florida, P.O. Box 100244, Gainesville, FL 32610, USA.
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL, USA; Department of Periodontology, College of Dentistry, University of Florida, P.O. Box 100434, Gainesville, FL 32610-0434, USA.
| |
Collapse
|
33
|
Huang FC. Differential regulation of interleukin-8 and human beta-defensin 2 in Pseudomonas aeruginosa-infected intestinal epithelial cells. BMC Microbiol 2014; 14:275. [PMID: 25433669 PMCID: PMC4261737 DOI: 10.1186/s12866-014-0275-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/24/2014] [Indexed: 01/05/2023] Open
Abstract
Background The human opportunistic pathogen, Pseudomonas aeruginosa (P. aeruginosa) carries the highest case fatality rate of all gram-negative infections. Unfortunately, antimicrobial therapy has not been demonstrated to improve clinical outcome and the emergence of multidrug resistant P. aeruginosa has become a major concern in the hospital setting. Fever and diarrhea are the two most common initial symptoms in P. aeruginosa sepsis in previously healthy infants and children. This implies that intestinal epithelial cells in first contact with the pathogen may play an important role in innate immunity to P. aeruginosa infection. Human beta–defensins-2 (hBD-2) and interleukin-8 (IL-8) are crucial for host defense at mucosa but IL-8 may give rise to characteristic pathology of colitis. Results Pseudomonas aeruginosa strain PAO1 was used to infect SW480, an intestinal epithelial cell. IL-8 and hBD-2 mRNA expression and protein secretion were then assessed in SW480 cells using RT-PCR and enzyme-linked immunosorbent assay (ELISA), respectively. Intracellular signaling pathways and nucleotide-binding oligomerization domain (NOD) 1 protein expression were analyzed by Western blot in SW480 cells in the presence or absence of inhibitors or transfected with siRNA. We demonstrate that prolonged infection by P. aeruginosa results in suppression of IL-8 but enhancement of hBD-2, either protein secretion and mRNA expression, in SW480 cells. Inhibitors of ERK suppressed but inhibitor of PI3K enhanced P. aeruginosa-induced IL-8 mRNA expression in SW480 cells while both signaling had no effect on P. aeruginosa-induced hBD-2 expression in SW480 cells. On the other hand, NOD 1 was illustrated to get involved in P. aeruginosa-induced hBD-2 mRNA expression and protein production in SW480 cells. Conclusions The P. aeruginosa-induced antimicrobial peptide in IECs continuously protect the host against prolonged infection, while modulation of proinflammatory responses prevents the host from the detrimental effects of overwhelming inflammation. Thus, P. aeruginosa-induced innate immunity in IECs represents a host protective mechanism, which may provide new insight into the pathogenesis of inflammatory bowel diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0275-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123, Ta-pei Road, Niao-sung District, Kaohsiung, Taiwan.
| |
Collapse
|
34
|
Mukherjee S, Biswas T. Activation of TOLLIP by porin prevents TLR2-associated IFN-γ and TNF-α-induced apoptosis of intestinal epithelial cells. Cell Signal 2014; 26:2674-82. [PMID: 25152369 DOI: 10.1016/j.cellsig.2014.08.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/15/2014] [Indexed: 11/25/2022]
Abstract
Interferon (IFN)-γ and tumor necrosis factor (TNF)-α cause chronic inflammation of the intestine leading to progression of inflammatory bowel disease (IBD), which is manifested through rapid apoptosis of the intestinal epithelial cells (iECs). Here, we show inhibition of IFN-γ and TNF-α-induced apoptosis of INT-407 cells by porin, a microbe-associated molecular pattern (MAMP) with affinity for toll-like receptor (TLR)2 and commonly present in Gram-negative bacteria. Proinflammatory cytokines induce apoptosis by activation of caspase 8 that triggers caspase 9 through Bax finally leading to activation of caspase 3, the executioner caspase. Interestingly, while IFN-γ and TNF-α promotes Bax expression, in contrast porin up-regulates anti-apoptotic Bcl-xL resulting in iEC survivability. We show elevated expression of TLR2 is a key requisite for IFN-γ and TNF-α mediated caspase 8 up-regulation that contributes to apoptosis of iECs. Down-regulation of TLR2 expression is central for checking apoptosis which is achieved by elevated level of toll-interacting protein (TOLLIP) in presence of porin. Attempts to limit IBD is in progress with anti-IFN-γ and anti-TNF-α Abs or use of IL-10. Although probiotic bacterial proteins have shown to successfully reduce IFN-γ and TNF-α mediated apoptosis, the exact mechanism of their action has remained elusive. This study identifies the underlying sequential events of transient TLR2 stimulation followed by its blocking in response to the bacterial outer membrane protein, which advocates intervention at TLR-juncture is crucial for controlling IBD.
Collapse
Affiliation(s)
- Subhadeep Mukherjee
- Division of Immunology, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Kolkata 700 010, West Bengal, India
| | - Tapas Biswas
- Division of Immunology, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Kolkata 700 010, West Bengal, India.
| |
Collapse
|
35
|
Gustafsson JK, Lindén SK, Alwan AH, Scholte BJ, Hansson GC, Sjövall H. Carbachol-induced colonic mucus formation requires transport via NKCC1, K⁺ channels and CFTR. Pflugers Arch 2014; 467:1403-1415. [PMID: 25139191 DOI: 10.1007/s00424-014-1595-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 08/01/2014] [Accepted: 08/06/2014] [Indexed: 12/26/2022]
Abstract
The colonic mucosa protects itself from the luminal content by secreting mucus that keeps the bacteria at a distance from the epithelium. For this barrier to be effective, the mucus has to be constantly replenished which involves exocytosis and expansion of the secreted mucins. Mechanisms involved in regulation of mucus exocytosis and expansion are poorly understood, and the aim of this study was to investigate whether epithelial anion secretion regulates mucus formation in the colon. The muscarinic agonist carbachol was used to induce parallel secretion of anions and mucus, and by using established inhibitors of ion transport, we studied how inhibition of epithelial transport affected mucus formation in mouse colon. Anion secretion and mucin exocytosis were measured by changes in membrane current and epithelial capacitance, respectively. Mucus thickness measurements were used to determine the carbachol effect on mucus growth. The results showed that the carbachol-induced increase in membrane current was dependent on NKCC1 co-transport, basolateral K(+) channels and Cftr activity. In contrast, the carbachol-induced increase in capacitance was partially dependent on NKCC1 and K(+) channel activity, but did not require Cftr activity. Carbachol also induced an increase in mucus thickness that was inhibited by the NKCC1 blocker bumetanide. However, mice that lacked a functional Cftr channel did not respond to carbachol with an increase in mucus thickness, suggesting that carbachol-induced mucin expansion requires Cftr channel activity. In conclusion, these findings suggest that colonic epithelial transport regulates mucus formation by affecting both exocytosis and expansion of the mucin molecules.
Collapse
Affiliation(s)
- Jenny K Gustafsson
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden.
| | - Sara K Lindén
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Ala H Alwan
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Bob J Scholte
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Gunnar C Hansson
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Henrik Sjövall
- Department of Internal Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Zhang B, Shao Y, Liu D, Yin P, Guo Y, Yuan J. Zinc prevents Salmonella enterica serovar Typhimurium-induced loss of intestinal mucosal barrier function in broiler chickens. Avian Pathol 2014; 41:361-7. [PMID: 22834550 DOI: 10.1080/03079457.2012.692155] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The study was carried out to evaluate the beneficial effects of supplemental zinc (Zn) on the intestinal mucosal barrier function in Salmonella enterica serovar Typhimurium-challenged broiler chickens in a 42-day experiment. A total of 336 1-day-old male Arbor Acres broiler chicks were assigned to eight treatment groups. A 4×2 factorial arrangement of treatments was used in a completely randomized experimental design to study the effects of levels of supplemental Zn (0, 40, 80 and 120 mg/kg diet), pathogen challenge (with or without S. Typhimurium challenge), and their interactions. S. Typhimurium infection caused reduction of growth performance (P<0.05) and intestinal injury, as determined by reduced (P<0.05) villus height/crypt depth ratio and sucrase activity in the ileum, increased (P<0.05) plasma endotoxin levels, and reduced (P<0.05) claudin-1, occludin and mucin-2 mRNA expression in the ileum at day 21. Zn pre-treatment tended to improve body weight gain (P=0.072) in the starter period, to increase the activity of ileal sucrase (P=0.077), to reduce plasma endotoxin levels (P=0.080), and to significantly increase (P<0.05) the villus height/crypt depth ratio and mRNA levels of occludin and claudin-1 in the ileum at day 21. The results indicated that dietary Zn supplementation appeared to alleviate the loss of intestinal mucosal barrier function induced by S. Typhimurium challenge and the partial mechanism might be related to the increased expression of occludin and claudin-1 in broiler chickens.
Collapse
Affiliation(s)
- Bingkun Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Key Laboratory of Feed Safety and Bioavailability, Ministry of Agriculture, Beijing 100193, PR China
| | | | | | | | | | | |
Collapse
|
37
|
Silva RC, Padovan ACB, Pimenta DC, Ferreira RC, da Silva CV, Briones MRS. Extracellular enolase of Candida albicans is involved in colonization of mammalian intestinal epithelium. Front Cell Infect Microbiol 2014; 4:66. [PMID: 24918088 PMCID: PMC4042164 DOI: 10.3389/fcimb.2014.00066] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/06/2014] [Indexed: 11/13/2022] Open
Abstract
Enolase is secreted by Candida albicans and is present in its biofilms although its extracellular function is unknown. Here we show that extracellular enolase mediates the colonization of small intestine mucosa by C. albicans. Assays using intestinal mucosa disks show that C. albicans adhesion is inhibited, in a dose dependent mode, either by pretreatment of intestinal epithelium mucosa disks with recombinant C. albicans enolase (70% at 0.5 mg/ml enolase) or by pretreatment of C. albicans yeasts with anti-enolase antibodies (48% with 20 μg antiserum). Also using flow cytometry, immunoblots of conditioned media and confocal microscopy we demonstrate that enolase is present in biofilms and that the extracellular enolase is not an artifact due to cell lysis, but must represent functional secretion of a stable form. This is the first direct evidence that C. albicans' extracellular enolase mediates colonization on its primary translocation site. Also, because enolase is encoded by a single locus in C. albicans, its dual role peptide, as glycolytic enzyme and extracellular peptide, is a remarkable example of gene sharing in fungi.
Collapse
Affiliation(s)
- Richard C Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Ana Carolina B Padovan
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo São Paulo, Brazil ; Disciplina de Infectologia, DMED, Universidade Federal de São Paulo São Paulo, Brazil
| | - Daniel C Pimenta
- Laboratório de Bioquímica e Biofísica, Instituto Butantã São Paulo, Brazil
| | - Renata C Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo São Paulo, Brazil ; Disciplina de Infectologia, DMED, Universidade Federal de São Paulo São Paulo, Brazil
| | - Claudio V da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo São Paulo, Brazil ; Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Marcelo R S Briones
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
38
|
Nieto-Pelegrin E, Meiler E, Martín-Villa JM, Benito-León M, Martinez-Quiles N. Crk adaptors negatively regulate actin polymerization in pedestals formed by enteropathogenic Escherichia coli (EPEC) by binding to Tir effector. PLoS Pathog 2014; 10:e1004022. [PMID: 24675776 PMCID: PMC3968158 DOI: 10.1371/journal.ppat.1004022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 02/05/2014] [Indexed: 01/04/2023] Open
Abstract
Infections by enteropathogenic Escherichia coli (EPEC) cause diarrhea linked to high infant mortality in developing countries. EPEC adheres to epithelial cells and induces the formation of actin pedestals. Actin polymerization is driven fundamentally through signaling mediated by Tir bacterial effector protein, which inserts in the plasma membrane of the infected cell. Tir binds Nck adaptor proteins, which in turn recruit and activate N-WASP, a ubiquitous member of the Wiskott-Aldrich syndrome family of proteins. N-WASP activates the Arp2/3 complex to promote actin polymerization. Other proteins aside from components of the Tir-Nck-N-WASP pathway are recruited to the pedestals but their functions are unknown. Here we investigate the function of two alternatively spliced isoforms of Crk adaptors (CrkI/II) and the paralog protein CrkL during pedestal formation by EPEC. We found that the Crk isoforms act as redundant inhibitors of pedestal formation. The SH2 domain of CrkII and CrkL binds to phosphorylated tyrosine 474 of Tir and competes with Nck to bind Tir, preventing its recruitment to pedestals and thereby inhibiting actin polymerization. EPEC infection induces phosphorylation of the major regulatory tyrosine in CrkII and CrkL, possibly preventing the SH2 domain of these proteins from interacting with Tir. Phosphorylated CrkII and CrkL proteins localize specifically to the plasma membrane in contact with EPEC. Our study uncovers a novel role for Crk adaptors at pedestals, opening a new perspective in how these oncoproteins regulate actin polymerization.
Collapse
Affiliation(s)
- Elvira Nieto-Pelegrin
- Department of Microbiology, School of Pharmacy, Complutense University, Madrid, Spain
| | - Eugenia Meiler
- Division of Immunology, School of Medicine, Complutense University, Madrid, Spain
| | | | - María Benito-León
- Division of Immunology, School of Medicine, Complutense University, Madrid, Spain
| | - Narcisa Martinez-Quiles
- Department of Microbiology, School of Pharmacy, Complutense University, Madrid, Spain
- Division of Immunology, School of Medicine, Complutense University, Madrid, Spain
- * E-mail:
| |
Collapse
|
39
|
Vásquez Cachay ME, Gomez EP, Rodríguez Gutiérrez JL, Lira Mejía B, Pérez NF, Zanuzzi CN, Barbeito C. Paneth Cell Identification in the Small Intestine of Guinea Pig Offsprings (Cavia porcellus). Anat Rec (Hoboken) 2014; 297:856-63. [DOI: 10.1002/ar.22890] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 01/08/2014] [Indexed: 01/21/2023]
Affiliation(s)
- María Elith Vásquez Cachay
- Animal Physiology Laboratory; Veterinary Medicine School; National University of San Marcos; San Borja Lima Perú
| | - Erika Pebe Gomez
- Animal Physiology Laboratory; Veterinary Medicine School; National University of San Marcos; San Borja Lima Perú
| | - José Luis Rodríguez Gutiérrez
- Experimental Centre IVITA-El Mantaro; Veterinary Medicine School; National University of San Marcos; San Borja Lima Perú
| | - Boris Lira Mejía
- Animal Physiology Laboratory; Veterinary Medicine School; National University of San Marcos; San Borja Lima Perú
| | - Néstor Falcón Pérez
- Veterinary and Zootecnic Medicine School; Cayetano Heredia University (UPCH); San Martin de Porres Lima Perú
| | - Carolina Natalia Zanuzzi
- Department of Histology and Embryology; School of Veterinary Sciences, National University of La Plata; La Plata Buenos Aires Argentina
- National Scientific and Technical Research Council (CONICET-CCT La Plata); Buenos Aires Argentina
| | - Claudio Barbeito
- Department of Histology and Embryology; School of Veterinary Sciences, National University of La Plata; La Plata Buenos Aires Argentina
- National Scientific and Technical Research Council (CONICET-CCT La Plata); Buenos Aires Argentina
| |
Collapse
|
40
|
Bacterial ligand stimulates TLR2-dependent chemokines of colon cell. Immunobiology 2014; 219:350-6. [PMID: 24565410 DOI: 10.1016/j.imbio.2013.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/02/2013] [Accepted: 12/28/2013] [Indexed: 01/05/2023]
Abstract
Shigella spp. are known to penetrate the colonic epithelial cells causing shigellosis, which results in production of convalescent antibodies against porin, the surface exposed major outer membrane protein. Porin has been categorized as primarily TLR2-ligand and here we validated its signaling procedure in colonic INT-407 cells simulating the host scenario. Porin up-regulated TLR2 and -6 followed by TLR2·MYD88 complex formation suggesting direct involvement of MYD88 for downstream signaling. Translocation of NF-κB p65 and p50 subunits on to the nucleus indicates involvement of the transcription factor in signaling. Porin-induced TLR signaling specifically stimulated the pro-inflammatory chemokine panel comprising of MIP-1α, MCP-1 and IL-8. Inhibition studies of TLR2 and NF-κB led to abrogation of the pro-inflammatory chemokine response, showing TLR-dependent signaling through NF-κB regulate gut activity. This work elucidates TLR2 not only scans pathogen-associated molecule but also has a direct role in maneuvering colon cell response.
Collapse
|
41
|
Abstract
Histone H1 is located at the inter-nucleosome and more correctly at both ends of the double-stranded DNA that protrude from the nucleosome unit. It has long been recognized to be localized only inside the nuclei as a constituent for packaging nucleosome into chromatin. Thus, it could be hardly believed that detatched or solubilized histone H1 plays the role of a host defense molecule. Given the old reports on histone-like basic proteins that show bacteriostatic functions, I herein chose some recent related articles and tried review them. Recent advances in research on the cell death mechanism makes it possible to understand that programmed cell death, (i.e. apoptosis) could serve as a good source of soluble histones. Some forms of them are highly probable to be bacteriostatic.
Collapse
Affiliation(s)
- Yoshiyuki Kanai
- Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
42
|
Bar-Yoseph F, Lifshitz Y, Cohen T. Review of sn-2 palmitate oil implications for infant health. Prostaglandins Leukot Essent Fatty Acids 2013; 89:139-43. [PMID: 23541258 DOI: 10.1016/j.plefa.2013.03.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human milk provides the optimal balanced nutrition for the growing infant in the first months after birth. The human mammary gland has evolved with unusual pathways, resulting in a specific positioning of fatty acids at the outer sn-1 and sn-3, and center sn-2 of the triacylglyceride, which is different from the triglycerides in other human tissues and plasma. The development of structured triglycerides enables mimicking the composition as well as structure of human milk fat in infant formulas. Studies conducted two decades ago, together with very recent studies, have provided increasing evidence that this unusual positioning of 16:0 in human milk triglycerides has a significant role for infant health in different directions, such as fat and calcium absorption, bone health, intestinal flora and infant comfort. This review aims to unravel the relevance of human milk triglyceride sn-2 16:0 for intestinal health and inflammatory pathways and for other post-absorption effects.
Collapse
|
43
|
Antimicrobial activity of high-mobility-group box 2: a new function to a well-known protein. Antimicrob Agents Chemother 2013; 57:4782-93. [PMID: 23877675 DOI: 10.1128/aac.00805-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The human intestinal tract is highly colonized by a vast number of microorganisms. Despite this permanent challenge, infections remain rare, due to a very effective barrier defense system. Essential effectors of this system are antimicrobial peptides and proteins (AMPs), which are secreted by intestinal epithelial and lymphoid cells, balance the gut microbial community, and prevent the translocation of microorganisms. Several antimicrobial proteins have already been identified in the gut. Nonetheless, we hypothesized that additional AMPs are yet to be discovered in this setting. Using biological screening based on antimicrobial function, here we identified competent antibacterial activity of high-mobility-group box 2 (HMGB2) against Escherichia coli. By recombinant expression, we confirmed this biologically new antimicrobial activity against different commensal and pathogenic bacteria. In addition, we demonstrated that the two DNA-binding domains (HMG boxes A and B) are crucial for the antibiotic function. We detected HMGB2 in several gastrointestinal tissues by mRNA analysis and immunohistochemical staining. In addition to the nuclei, we also observed HMGB2 in the cytoplasm of intestinal epithelial cells. Furthermore, HMGB2 was detectable in vitro in the supernatants of two different cell types, supporting an extracellular function. HMGB2 expression was not changed in inflammatory bowel disease but was detected in certain stool samples of patients, whereas it was absent from control individuals. Taken together, we characterized HMGB2 as an antimicrobial protein in intestinal tissue, complementing the diverse repertoire of gut mucosal defense molecules.
Collapse
|
44
|
Lu P, Bar-Yoseph F, Levi L, Lifshitz Y, Witte-Bouma J, de Bruijn ACJM, Korteland-van Male AM, van Goudoever JB, Renes IB. High beta-palmitate fat controls the intestinal inflammatory response and limits intestinal damage in mucin Muc2 deficient mice. PLoS One 2013; 8:e65878. [PMID: 23776564 PMCID: PMC3680492 DOI: 10.1371/journal.pone.0065878] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/29/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Palmitic-acid esterified to the sn-1,3 positions of the glycerol backbone (alpha, alpha'-palmitate), the predominant palmitate conformation in regular infant formula fat, is poorly absorbed and might cause abdominal discomfort. In contrast, palmitic-acid esterified to the sn-2 position (beta-palmitate), the main palmitate conformation in human milk fat, is well absorbed. The aim of the present study was to examine the influence of high alpha, alpha'-palmitate fat (HAPF) diet and high beta-palmitate fat (HBPF) diet on colitis development in Muc2 deficient (Muc2(-/-)) mice, a well-described animal model for spontaneous enterocolitis due to the lack of a protective mucus layer. METHODS Muc2(-/-) mice received AIN-93G reference diet, HAPF diet or HBPF diet for 5 weeks after weaning. Clinical symptoms, intestinal morphology and inflammation in the distal colon were analyzed. RESULTS Both HBPF diet and AIN-93G diet limited the extent of intestinal erosions and morphological damage in Muc2(-/-) mice compared with HAPF diet. In addition, the immunosuppressive regulatory T (Treg) cell response as demonstrated by the up-regulation of Foxp3, Tgfb1 and Ebi3 gene expression levels was enhanced by HBPF diet compared with AIN-93G and HAPF diets. HBPF diet also increased the gene expression of Pparg and enzymatic antioxidants (Sod1, Sod3 and Gpx1), genes all reported to be involved in promoting an immunosuppressive Treg cell response and to protect against colitis. CONCLUSIONS This study shows for the first time that HBPF diet limits the intestinal mucosal damage and controls the inflammatory response in Muc2(-/-) mice by inducing an immunosuppressive Treg cell response.
Collapse
Affiliation(s)
- Peng Lu
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, the Netherlands
| | | | | | | | - Janneke Witte-Bouma
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | | | | | - Johannes B. van Goudoever
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, the Netherlands
- Department of Pediatrics, VU University Medical Center, Amsterdam, the Netherlands
| | - Ingrid B. Renes
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
45
|
Lan D, Tang C, Yue H, Sun H, Cui L, Hua X, Li J. Microarray analysis of differentially expressed transcripts in porcine intestinal epithelial cells (IPEC-J2) infected with porcine sapelovirus as a model to study innate immune responses to enteric viruses. Arch Virol 2013; 158:1467-75. [PMID: 23417395 DOI: 10.1007/s00705-013-1638-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/09/2013] [Indexed: 12/25/2022]
Abstract
The local intestinal mucosa, the largest mucosal immune system in animals, plays an important role in resistance against intestinal pathogen infection. However, the molecular antiviral mechanisms of the intestinal mucosa remain poorly understood. In this study, we screened and identified differentially expressed transcripts in (PSV) porcine intestinal epithelial cells (IPEC-J2) infected with porcine sapelovirus using microarray analysis. A total of 2298 differentially expressed genes were screened at four time points during PSV infection. These genes were involved in numerous physical systems and molecular pathways, and particularly, some innate immune-associated pathways were significant. The results showed that large amounts of type I interferon were induced, and the related interferon effect pathway was activated when IPEC-J2 cells were infected with PSV. Three pathways of innate immune receptors, including Toll-like, NOD-like, and RIG-I-like receptors, were also activated. The antigen was then processed and presented through the MHCI and MHCII pathways. Interestingly, we found that the secretion network of IgA was activated in the early stage of PSV infection. Two exogenous and endogenous apoptosis pathways were also activated during PSV infection. The results revealed changes in gene transcription, particularly those of innate immune pathway genes that were associated with PSV infection in IPEC-J2 cells.
Collapse
Affiliation(s)
- Daoliang Lan
- Hi-tech Research and Development Base for Qinghai-Tibet Plateau Ecological Conservation and Stock Farming, Southwest University for Nationality, Chengdu, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Arciero J, Bard Ermentrout G, Siggers R, Afrazi A, Hackam D, Vodovotz Y, Rubin J. Modeling the interactions of bacteria and Toll-like receptor-mediated inflammation in necrotizing enterocolitis. J Theor Biol 2012; 321:83-99. [PMID: 23238281 DOI: 10.1016/j.jtbi.2012.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 11/19/2012] [Accepted: 12/03/2012] [Indexed: 01/18/2023]
Abstract
Necrotizing enterocolitis (NEC) is a severe disease of the gastrointestinal tract in premature infants, characterized by a disrupted intestinal epithelium and an exaggerated pro-inflammatory response. Since the activation of Toll-like receptor-4 (TLR4) blocks cell migration and proliferation and contributes to an uncontrolled inflammatory response within the intestine, this receptor has been identified as a key contributor to the development of NEC. Toll-like receptor-9 (TLR9) has been shown to sense bacterial genome components (CpG DNA) and to play an anti-inflammatory role in NEC. We present in vitro results demonstrating direct inhibition of TLR4 activation by CpG DNA, and we develop a mathematical model of bacteria-immune interactions within the intestine to investigate how such inhibition of TLR4 signaling might alter inflammation, associated bacterial invasion of tissue, and resulting outcomes. The model predicts that TLR9 can inhibit both the beneficial and detrimental effects of TLR4, and thus a proper balance of action by these two receptors is needed to promote intestinal health. The model results are also used to explore three interventions that could potentially prevent the development of NEC: reducing bacteria in the mucus layer, administering probiotic treatment, and blocking TLR4 activation. While the model shows that these interventions would be successful in most cases, the model is also used to identify situations in which the proposed treatments might be harmful.
Collapse
Affiliation(s)
- Julia Arciero
- Department of Mathematics, IUPUI, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Cetinkaya M, Cansev M, Cekmez F, Tayman C, Canpolat FE, Kafa IM, Uysal S, Tunc T, Sarici SU. CDP-choline reduces severity of intestinal injury in a neonatal rat model of necrotizing enterocolitis. J Surg Res 2012; 183:119-28. [PMID: 23228325 DOI: 10.1016/j.jss.2012.11.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 10/03/2012] [Accepted: 11/16/2012] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cytidine 5'-diphosphocholine (CDP-choline) is an endogenous intermediate in the biosynthesis of phosphatidylcholine, a contributor to the mucosal defense of the intestine. The aim of this study was to evaluate the possible cytoprotective effect of CDP-choline treatment on intestinal cell damage, membrane phospholipid content, inflammation, and apoptosis in a neonatal rat model of necrotizing enterocolitis (NEC). METHODS We divided a total of 30 newborn pups into three groups: control, NEC, and NEC + CDP-choline. We induced NEC by enteral formula feeding, exposure to hypoxia-hyperoxia, and cold stress. We administered CDP-choline intraperitoneally at 300 mg/kg/d for 3 d starting from the first day of life. We evaluated apoptosis macroscopically and histopathologically in combination with proinflammatory cytokines in the gut samples. Moreover, we determined membrane phospholipid levels as well as activities of xanthine oxidase, superoxide dismutase, glutathione peroxidase, and myeloperoxidase enzymes and the malondialdehyde content of intestinal tissue. RESULTS Mean clinical sickness score, macroscopic gut assessment score, and intestinal injury score were significantly improved, whereas mean apoptosis score and caspase-3 levels were significantly reduced in pups in the NEC + CDP-choline group compared with the NEC group. Tissue proinflammatory cytokine (interleukin-1β, interleukin-6, and tumor necrosis factor-α) levels as well as tissue malondialdehyde content and myeloperoxidase activities were reduced, whereas glutathione peroxidase and superoxide dismutase activities were preserved in the NEC + CDP-choline group. In addition, NEC damage reduced intestinal tissue membrane phospholipids, whereas CDP-choline significantly enhanced total phospholipid and phosphatidylcholine levels. Long-term follow-up in additional experiments revealed increased body weight, decreased clinical sickness scores, and enhanced survival in CDP-choline-receiving versus saline-receiving pups with NEC lesions. CONCLUSIONS Our study reports, for the first time, beneficial effects of CDP-choline treatment on intestinal injury in a neonatal rat model of NEC. Our data suggest that CDP-choline may be used as an effective therapeutic agent to prevent NEC.
Collapse
Affiliation(s)
- Merih Cetinkaya
- Division of Neonatology, Department of Pediatrics, Gulhane Military Medical Academy, Ankara, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Saei AA, Barzegari A. The microbiome: the forgotten organ of the astronaut’s body – probiotics beyond terrestrial limits. Future Microbiol 2012; 7:1037-46. [DOI: 10.2217/fmb.12.82] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Space medicine research has drawn immense attention toward provision of efficient life support systems during long-term missions into space. However, in extended missions, a wide range of diseases may affect astronauts. In space medicine research, the gastrointestinal microbiome and its role in maintaining astronauts’ health has received little attention. We would like to draw researchers’ attention to the significant role of microbiota. Because of the high number of microorganisms in the human body, man has been called a ‘supra-organism’ and gastrointestinal flora has been referred to as ‘a virtual organ of the human body’. In space, the lifestyle, sterility of spaceship and environmental stresses can result in alterations in intestinal microbiota, which can lead to an impaired immunity and predispose astronauts to illness. This concern is heightened by increase in virulence of pathogens in microgravity. Thus, design of a personal probiotic kit is recommended to improve the health status of astronauts.
Collapse
Affiliation(s)
- Amir Ata Saei
- Research Center for Pharmaceutical Nanotechnology, Astrobiology & Space Medicine Laboratory, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Astrobiology & Space Medicine Laboratory, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
François M, Birman E, Forner KA, Gaboury L, Galipeau J. Adoptive transfer of mesenchymal stromal cells accelerates intestinal epithelium recovery of irradiated mice in an interleukin-6-dependent manner. Cytotherapy 2012; 14:1164-70. [PMID: 22574720 DOI: 10.3109/14653249.2012.684378] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AIMS Apoptosis of radiosensitive cells in the bone marrow and gut is a serious, at times life-threatening, complication arising from radiation exposure. METHODS We investigated whether adoptive transfer of allogeneic bone marrow-derived mesenchymal stromal cells (MSC) could exert cytoprotective and life-sparing effects in a mouse model of sublethal total body irradiation (TBI). RESULTS We demonstrated that a single intraperitoneal injection of C57Bl/6 MSC given to major histocompatibility complex (MHC)-mismatched Balb/c mice within 24 h of sublethal TBI significantly reduced mortality in a dose-dependent manner. Histologic analysis and Ki67 immunostaining of jejunum sections collected 3 and 6 days post-TBI indicated that MSC protected the gastrointestinal epithelium from TBI-induced damage and significantly accelerated recovery of the gut by stimulating proliferation of the crypt cell pool. Using interleukin-6(-/-) (IL-6) MSC, we demonstrated that IL-6 expressed by MSC played a role in gastrointestinal epithelium regeneration. CONCLUSIONS Our results suggest that allogeneic MHC-mismatched MSC may be exploited to reduce gastrointestinal complications and mortality arising from ionizing radiation exposure.
Collapse
Affiliation(s)
- Moïra François
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | | | | | | | | |
Collapse
|
50
|
Wos-Oxley ML, Bleich A, Oxley AP, Kahl S, Janus LM, Smoczek A, Nahrstedt H, Pils MC, Taudien S, Platzer M, Hedrich HJ, Medina E, Pieper DH. Comparative evaluation of establishing a human gut microbial community within rodent models. Gut Microbes 2012; 3:234-49. [PMID: 22572831 PMCID: PMC3427216 DOI: 10.4161/gmic.19934] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The structure of the human gut microbial community is determined by host genetics and environmental factors, where alterations in its structure have been associated with the onset of different diseases. Establishing a defined human gut microbial community within inbred rodent models provides a means to study microbial-related pathologies, however, an in-depth comparison of the established human gut microbiota in the different models is lacking. We compared the efficiency of establishing the bacterial component of a defined human microbial community within germ-free (GF) rats, GF mice, and antibiotic-treated specific pathogen-free mice. Remarkable differences were observed between the different rodent models. While the majority of abundant human-donor bacterial phylotypes were established in the GF rats, only a subset was present in the GF mice. Despite the fact that members of the phylum Bacteriodetes were well established in all rodent models, mice enriched for phylotypes related to species of Bacteroides. In contrary to the efficiency of Clostridiales to populate the GF rat in relative proportions to that of the human-donor, members of Clostridia cluster IV only poorly colonize the mouse gut. Thus, the genetic background of the different recipient rodent systems (that is, rats and mice) strongly influences the nature of the populating human gut microbiota, determining each model's biological suitability.
Collapse
Affiliation(s)
- Melissa L. Wos-Oxley
- Microbial Interactions and Processes Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany,Correspondence to: Melissa L. Wos-Oxley,
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility; Hannover Medical School; Hannover, Germany
| | - Andrew P.A. Oxley
- Infection Immunology Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany
| | - Silke Kahl
- Microbial Interactions and Processes Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany
| | - Lydia M. Janus
- Institute for Laboratory Animal Science and Central Animal Facility; Hannover Medical School; Hannover, Germany
| | - Anna Smoczek
- Institute for Laboratory Animal Science and Central Animal Facility; Hannover Medical School; Hannover, Germany
| | - Hannes Nahrstedt
- Microbial Interactions and Processes Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany
| | - Marina C. Pils
- Central Animal Facility; Helmholtz Centre for Infection Research; Braunschweig, Germany
| | - Stefan Taudien
- Genome Analysis; Leibniz Institute for Age Research; Fritz Lipmann Institute; Jena, Germany
| | - Matthias Platzer
- Genome Analysis; Leibniz Institute for Age Research; Fritz Lipmann Institute; Jena, Germany
| | - Hans-Jürgen Hedrich
- Institute for Laboratory Animal Science and Central Animal Facility; Hannover Medical School; Hannover, Germany
| | - Eva Medina
- Infection Immunology Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany
| | - Dietmar H. Pieper
- Microbial Interactions and Processes Research Group; Department of Medical Microbiology; Helmholtz Centre for Infection Research; Braunschweig, Germany
| |
Collapse
|