1
|
Zhang Y, Tian Y, Zhong X, Zhang R, Yang S, Jin J, Lyu C, Fan J, Shi B, Zhu K, Xiao Y, Lin N, Ma D, Tou J, Shu Q, Lai D. RNF31-mediated IKKα ubiquitination aggravates inflammation and intestinal injury through regulating NF-κB activation in human and mouse neonates. Life Sci 2024; 352:122893. [PMID: 38971367 DOI: 10.1016/j.lfs.2024.122893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
AIMS Neonatal necrotizing enterocolitis (NEC) is a leading cause of intestine inflammatory disease, and macrophage is significantly activated during NEC development. Posttranslational modifications (PTMs) of proteins, particularly ubiquitination, play critical roles in immune response. This study aimed to investigate the effects of ubiquitin-modified proteins on macrophage activation and NEC, and discover novel NEC-related inflammatory proteins. MATERIALS AND METHODS Proteomic and ubiquitin proteomic analyses of intestinal macrophages in NEC/healthy mouse pups were carried out. In vitro macrophage inflammation model and in vivo NEC mouse model, as well as clinical human samples were used for further verification the inhibitor of nuclear factor-κB kinase α (IKKα) ubiquitination on NEC development through Western blot, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR) and flow cytometry. KEY FINDINGS We report here that IKKα was a new ubiquitin-modified protein during NEC through ubiquitin proteomics, and RING finger protein 31 (RNF31) acted as an E3 ligase to be involved in IKKα degradation. Inhibition of IKKα ubiquitination and degradation with siRNF31 or proteasome inhibitor decreased nuclear factor-κB (NF-κB) activation, thereby decreasing the expression of pro-inflammatory factors and M1 macrophage polarization, resulting in reliving the severity of NEC. SIGNIFICANCE Our study suggests the activation of RNF31-IKKα-NF-κB axis triggering NEC development and suppressing RNF31-mediated IKKα degradation may be therapeutic strategies to be developed for NEC treatment.
Collapse
Affiliation(s)
- Yuebai Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yangfan Tian
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ruoyang Zhang
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingyi Jin
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiajie Fan
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Bo Shi
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kun Zhu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Xiao
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Nan Lin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Daqing Ma
- Perioperative and Systems Medicine, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
2
|
Impairment in the Intestinal Morphology and in the Immunopositivity of Toll-like Receptor-4 and Other Proteins in an Autistic Mouse Model. Int J Mol Sci 2022; 23:ijms23158731. [PMID: 35955865 PMCID: PMC9369377 DOI: 10.3390/ijms23158731] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/03/2022] Open
Abstract
Autism spectrum disorder (ASD) identifies a neurodevelopmental disease defined by social impairments and repetitive or stereotyped behaviors. The etiology of ASD remains unclear; it primarily affects the brain, but a link between gastrointestinal (GI) diseases, inflammatory mucosal pathology and this disorder has been suggested. In particular, a central role seems to be played by an imbalance in pro-and anti-inflammatory cytokines, oxidative stress, and apoptosis. Toll-like receptor 4 (TLR4) is a protein of innate immunity responsible for the regulation and maintenance of intestinal homeostasis. Through histochemical and immunohistochemical evaluations we analyzed the intestinal morphology and the immunopositivity of TLR4 and of other pro-inflammatory and apoptotic proteins in BTBR T+Itpr3tf/J mice. Morphological data showed that the mucosal tunica presented longer intestinal villi. The length of the villi and the epithelial surface determine the exchanges of the intestinal mucosa with luminal contents, modifying the microbiota composition. The biochemical and immunohistochemical results indicated a close relationship among the increase of TLR4 and the activation of NF-kB subunits (p65 and p50) and pro-inflammatory and apoptotic proteins, such as cyclooxygenase-2, interleukin-1β, inducible nitric oxide synthase, tumor nuclear factor—alpha, caspase-3, caspase-8. These preliminary results require more in-depth study but they suggest the TLR4 signaling pathway as a possible target for therapeutic approaches to reduce GI disorders in ASD.
Collapse
|
3
|
Gomart A, Vallée A, Lecarpentier Y. Necrotizing Enterocolitis: LPS/TLR4-Induced Crosstalk Between Canonical TGF-β/Wnt/β-Catenin Pathways and PPARγ. Front Pediatr 2021; 9:713344. [PMID: 34712628 PMCID: PMC8547806 DOI: 10.3389/fped.2021.713344] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Necrotizing enterocolitis (NEC) represents one of the major causes of morbidity and mortality in premature infants. Several recent studies, however, have contributed to a better understanding of the pathophysiology of this dreadful disease. Numerous intracellular pathways play a key role in NEC, namely: bacterial lipopolysaccharide (LPS), LPS toll-like receptor 4 (TLR4), canonical Wnt/β-catenin signaling and PPARγ. In a large number of pathologies, canonical Wnt/β-catenin signaling and PPARγ operate in opposition to one another, so that when one of the two pathways is overexpressed the other is downregulated and vice-versa. In NEC, activation of TLR4 by LPS leads to downregulation of the canonical Wnt/β-catenin signaling and upregulation of PPARγ. This review aims to shed light on the complex intracellular mechanisms involved in this pathophysiological profile by examining additional pathways such as the GSK-3β, NF-κB, TGF-β/Smads, and PI3K-Akt pathways.
Collapse
Affiliation(s)
- Alexia Gomart
- Département de Pédiatrie et Médecine de l'adolescent, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Alexandre Vallée
- Department of Clinical Research and Innovation, Foch Hospital, Suresnes, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
4
|
Managlia E, Liu SXL, Yan X, Tan XD, Chou PM, Barrett TA, De Plaen IG. Blocking NF-κB Activation in Ly6c + Monocytes Attenuates Necrotizing Enterocolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:604-618. [PMID: 30593820 PMCID: PMC6412404 DOI: 10.1016/j.ajpath.2018.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease affecting premature infants with intestinal inflammation and necrosis. The neonatal intestinal inflammatory response is rich in macrophages, and blood monocyte count is low in human NEC. We previously found that NF-κB mediates the intestinal injury in experimental NEC. However, the role of NF-κB in myeloid cells during NEC remains unclear. Herein, inhibitor of kappaB kinase β (IKKβ), a critical kinase mediating NF-κB activation, was deleted in lysozyme M (Lysm)-expressing cells, which were found to be Cd11b+Ly6c+ monocytes but not Cd11b+Ly6c- macrophages in the dam-fed neonatal mouse intestine. NEC induced differentiation of monocytes into intestinal macrophages and up-regulation of monocyte recruitment genes (eg, L-selectin) in the macrophage compartment in wild-type mice, but not in pups with IKKβ deletion in Lysm+ cells. Thus, NF-κB is required for NEC-induced monocyte activation, recruitment, and differentiation in neonatal intestines. Furthermore, pups with Lysm-IKKβ deletion had improved survival and decreased incidence of severe NEC compared with littermate controls. Decreased NEC severity was not associated with an improved intestinal barrier. In contrast, NEC was unabated in mice with IKKβ deletion in intestinal epithelial cells. Together, these data suggest that recruitment of Ly6c+ monocytes into the intestine, NF-κB activation in these cells, and differentiation of Ly6c+ monocytes into macrophages are critical cellular and molecular events in NEC development to promote disease.
Collapse
Affiliation(s)
- Elizabeth Managlia
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Shirley X L Liu
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiaocai Yan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Xiao-Di Tan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Gastroenterology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Terrence A Barrett
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Isabelle G De Plaen
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
5
|
Natarajan K, Abraham P, Kota R, Isaac B. NF-κB-iNOS-COX2-TNF α inflammatory signaling pathway plays an important role in methotrexate induced small intestinal injury in rats. Food Chem Toxicol 2018; 118:766-783. [DOI: 10.1016/j.fct.2018.06.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/16/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022]
|
6
|
Wang X, Bu HF, Liu SXL, De Plaen IG, Tan XD. Molecular Mechanisms Underlying the Regulation of the MFG-E8 Gene Promoter Activity in Physiological and Inflammatory Conditions. J Cell Biochem 2016; 116:1867-79. [PMID: 25711369 DOI: 10.1002/jcb.25142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/17/2015] [Indexed: 12/15/2022]
Abstract
Milk fat globule-EGF factor 8 (MFG-E8) is expressed by macrophages and plays an important role in attenuating inflammation and maintaining tissue homeostasis. Previously, we and others found that lipopolysaccharide (LPS) inhibits MFG-E8 gene expression in macrophages. Here, we characterized the 5'-flanking region of the mouse MFG-E8 gene. To functionally analyze the upstream regulatory region of the MFG-E8 gene, a series of luciferase reporter gene constructs containing deleted or mutated regulatory elements were prepared. Using the luciferase assay, we revealed that Sp1 binding motifs within the proximal promoter region were necessary for full activity of the MFG-E8 promoter, whereas AP-1 like binding sequence at -372 played a role in governing the promoter activity at a homeostatic level. With chromatin immunoprecipitation assay, we showed that Sp1 and c-Jun physically interact with the MFG-E8 promoter region in vivo. In addition, Sp1 was found to regulate the MFG-E8 promoter activity positively and c-Jun negatively. Furthermore, we demonstrated that LPS inhibited MFG-E8 promoter activity via targeting Sp1 and AP-1-like motifs in the 5'-flanking region. Collectively, our data indicate that Sp1 and AP-1-related factors are involved in the regulation of MFG-E8 gene transcription by targeting their binding sites in the 5'-flanking region under physiological and inflammatory states.
Collapse
Affiliation(s)
- Xiao Wang
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Heng-Fu Bu
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Shirley X L Liu
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Isabelle G De Plaen
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao-Di Tan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Research & Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
7
|
Hunter CJ, De Plaen IG. Inflammatory signaling in NEC: Role of NF-κB, cytokines and other inflammatory mediators. ACTA ACUST UNITED AC 2013; 21:55-65. [PMID: 24388163 DOI: 10.1016/j.pathophys.2013.11.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Catherine J Hunter
- Department of Surgery, Division of Pediatric Surgery, Northwestern University Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, United States
| | - Isabelle G De Plaen
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Avenue, Box 45, Chicago, IL 60611, United States.
| |
Collapse
|
8
|
Borthakur A, Bhattacharyya S, Kumar A, Anbazhagan AN, Tobacman JK, Dudeja PK. Lactobacillus acidophilus alleviates platelet-activating factor-induced inflammatory responses in human intestinal epithelial cells. PLoS One 2013; 8:e75664. [PMID: 24130731 PMCID: PMC3794005 DOI: 10.1371/journal.pone.0075664] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/16/2013] [Indexed: 02/06/2023] Open
Abstract
Probiotics have been used as alternative prevention and therapy modalities in intestinal inflammatory disorders including inflammatory bowel diseases (IBD) and necrotizing enterocolitis (NEC). Pathophysiology of IBD and NEC includes the production of diverse lipid mediators, including platelet-activating factor (PAF) that mediate inflammatory responses in the disease. PAF is known to activate NF-κB, however, the mechanisms of PAF-induced inflammation are not fully defined. We have recently described a novel PAF-triggered pathway of NF-κB activation and IL-8 production in intestinal epithelial cells (IECs), requiring the pivotal role of the adaptor protein Bcl10 and its interactions with CARMA3 and MALT1. The current studies examined the potential role of the probiotic Lactobacillus acidophilus in reversing the PAF-induced, Bcl10-dependent NF-κB activation and IL-8 production in IECs. PAF treatment (5 µM×24 h) of NCM460 and Caco-2 cells significantly increased nuclear p65 NF-κB levels and IL-8 secretion (2-3-fold, P<0.05), compared to control, which were blocked by pretreatment of the cells for 6 h with L. acidophilus (LA) or its culture supernatant (CS), followed by continued treatments with PAF for 24 h. LA-CS also attenuated PAF-induced increase in Bcl10 mRNA and protein levels and Bcl10 promoter activity. LA-CS did not alter PAF-induced interaction of Bcl10 with CARMA3, but attenuated Bcl10 interaction with MALT1 and also PAF-induced ubiquitination of IKKγ. Efficacy of bacteria-free CS of LA in counteracting PAF-induced inflammatory cascade suggests that soluble factor(s) in the CS of LA mediate these effects. These results define a novel mechanism by which probiotics counteract PAF-induced inflammation in IECs.
Collapse
Affiliation(s)
- Alip Borthakur
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Sumit Bhattacharyya
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Arivarasu Natarajan Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Joanne K. Tobacman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Pradeep K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| |
Collapse
|
9
|
Abstract
The pathogenesis of necrotizing enterocolitis (NEC) is complex and its speed of progression is variable. To gain understanding of the disease, researchers have examined tissues resected from patients with NEC; however, as these are obtained at late stages of the disease, they do not yield clues about the early pathogenic events leading to NEC. Therefore, animal models are used and have helped identify a role for several mediators of the inflammatory network in NEC. In this article, we discuss the evidence for the role of these inflammatory mediators and conclude with a current unifying hypothesis regarding NEC pathogenesis.
Collapse
Affiliation(s)
- Isabelle G. De Plaen
- Associate Professor of Pediatrics, Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Children’s Hospital of Chicago Research Center, Ann and Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Box 45, Chicago, IL 60611-2605, U.S.A., Tel: (773)-755-6379; fax: (312)-227-9758
| |
Collapse
|
10
|
Kim EK, Lee KY, Lee HJ, Lee JA, Choi CW, Kim HS, Kim BI, Choi JH. Heat shock pretreatment reduces intestinal injury in a neonatal rat model of early necrotizing enterocolitis. Neonatology 2013; 103:1-6. [PMID: 22948204 DOI: 10.1159/000339179] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/30/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND Increased pro-inflammatory cytokines are suggested in the pathogenesis of necrotizing enterocolitis (NEC). The transcription factor, nuclear factor-ĸB (NF-ĸB), is a central regulator of inflammatory and immune responses, and recent rodent NEC models have shown that NF-ĸB may have a critical role in the disease processes that underlie NEC. Heat shock proteins have important functions in response to stress-related events in a variety of systems, including digestive organs. OBJECTIVES We investigated whether heat shock pretreatment protects intestinal epithelial damage in the early NEC rat model. METHODS We generated human NEC-like lesions in neonatal rat ileum by administering oral endotoxin (10 mg/kg), intermittent 8% hypoxia, and hypertonic formula. Heat shock was administered by raising the chamber temperature to 42°C for 20 min, 3 h prior to endotoxin ingestion. RESULTS Heat shock pretreatment increased the expression of HSP70 in the ileal tissue and attenuated histological severity of early experimental NEC. NF-ĸB was activated in the ileal tissue of the NEC group and this activation was attenuated by heat shock pretreatment, which was determined by electrophoretic mobility shift assay and Western blot analysis of p50 in subcellular fractionated samples. CONCLUSIONS Heat shock pretreatment reduced the incidence and severity of early experimental NEC in rats. A possible mechanism underlying this protective effect includes inhibition of NF-ĸB activation through increased HSP70 expression.
Collapse
Affiliation(s)
- Ee-Kyung Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Xu LF, Xu C, Mao ZQ, Teng X, Ma L, Sun M. Disruption of the F-actin cytoskeleton and monolayer barrier integrity induced by PAF and the protective effect of ITF on intestinal epithelium. Arch Pharm Res 2011; 34:245-51. [PMID: 21380808 DOI: 10.1007/s12272-011-0210-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 07/28/2010] [Accepted: 07/31/2010] [Indexed: 01/30/2023]
Abstract
To explore whether platelet-activating factor (PAF) can disrupt the intestinal epithelial barrier directly and is associated with structural alterations of the F-actin-based cytoskeleton, and to observe the protective effect of intestinal trefoil factor (ITF), we establish an intestinal epithelia barrier model using Caco-2 cells in vitro. Transepithelial electrical resistance and unidirectional flux of lucifer yellow were measured to evaluate barrier permeability; immunofluorescent staining and flow cytometry were applied to observe morphological alterations and to quantify proteins of the F-actin cytoskeleton: the tight junction marker ZO-1 and Claudin-1 were observed using immunofluorescent staining. PAF significantly increased paracellular permeability, at the same time, F-actin and tight junction proteins were disrupted. It was thought that ITF could reverse the high permeability by restoring normal F-actin, ZO-1 and Claudin-1 structures. These results collectively demonstrated that PAF plays an important role in the regulation of mucosal permeability and the effects of PAF are correlated with structural alterations of the F-actin-based cytoskeleton and of tight junctions. ITF can protect intestinal epithelium against PAF-induced disruption by restricting the rearrangement of the F-actin cytoskeleton and of tight junctions.
Collapse
Affiliation(s)
- Ling-fen Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
12
|
|
13
|
Borthakur A, Bhattacharyya S, Alrefai WA, Tobacman JK, Ramaswamy K, Dudeja PK. Platelet-activating factor-induced NF-kappaB activation and IL-8 production in intestinal epithelial cells are Bcl10-dependent. Inflamm Bowel Dis 2010; 16:593-603. [PMID: 19714753 PMCID: PMC3740729 DOI: 10.1002/ibd.21092] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Platelet-activating factor (PAF), a potent proinflammatory phospholipid mediator, has been implicated in inducing intestinal inflammation in diseases such as inflammatory bowel disease (IBD) and necrotizing enterocolitis (NEC). However, its mechanisms of inducing inflammatory responses are not fully understood. Therefore, studies were designed to explore the mechanisms of PAF-induced inflammatory cascade in intestinal epithelial cells. METHODS Nuclear factor kappa B (NF-kappaB) activation was measured by luciferase assay and enzyme-linked immunosorbent assay (ELISA), and interleukin 8 (IL-8) production was determined by ELISA. B-cell lymphoma 10 (Bcl10), caspase recruitment domain-containing membrane-associated guanylate kinase protein 3 (CARMA3), and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) mRNA and protein levels were assessed by real-time reverse-transcription polymerase chain reaction (RT-PCR) and Western blot, respectively. siRNA silencing of Bcl10 was used to examine its role in PAF-induced NF-kappaB activation and IL-8 production. The promoter region of the Bcl10 gene was cloned with the PCR method and promoter activity measured by luciferase assay. RESULTS The adaptor protein Bcl10 appeared to play an important role in the PAF-induced inflammatory pathway in human intestinal epithelial cells. Bcl10 was required for PAF-induced I kappaB alpha phosphorylation, NF-kappaB activation, and IL-8 production in NCM460, a cell line derived from normal human colon, and Caco-2, a transformed human intestinal cell line. PAF also stimulated Bcl10 interactions with CARMA3 and MALT1, and upregulated Bcl10 expression in these cells via transcriptional regulation. CONCLUSIONS These findings highlight a novel PAF-induced inflammatory pathway in intestinal epithelial cells, requiring Bcl10 as a critical mediator and involving CARMA3/Bcl10/MALT1 interactions. The proinflammatory effects of PAF play prominent roles in the pathogenesis of IBD and this pathway may present important targets for intervention in chronic inflammatory diseases of the intestine.
Collapse
Affiliation(s)
- Alip Borthakur
- Author to whom all correspondence including reprint requests should be addressed: Alip Borthakur, Ph.D., Department of Medicine, Section of Digestive Diseases and Nutrition, University of Illinois at Chicago, 840 South Wood Street (MC716), Chicago, IL 60612, , Phone: 312-569-7463, FAX: 312-569-7458
| | | | | | | | | | | |
Collapse
|
14
|
Liu SXL, Tian R, Baskind H, Hsueh W, De Plaen IG. Platelet-activating factor induces the processing of nuclear factor-kappaB p105 into p50, which mediates acute bowel injury in mice. Am J Physiol Gastrointest Liver Physiol 2009; 297:G76-81. [PMID: 19460845 PMCID: PMC2711750 DOI: 10.1152/ajpgi.00053.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Platelet-activating factor (PAF), an endogenous proinflammatory phospholipid, when injected intravascularly to rats and mice, causes shock, acute bowel injury, and a rapid activation of NF-kappaB p50-p50 with upregulation of the chemokine CXCL2 in the intestine. In this study, we investigate the mechanism of NF-kappaB activation and the role of the NF-kappaB p50 subunit in PAF-induced shock and acute bowel injury. NF-kappaB p50-deficient mice and wild-type mice were anesthetized and tracheotomized, and their carotid artery was cannulated for blood pressure monitoring, blood sampling, and PAF administration. For determination of bowel injury, shock, and survival, PAF (2.2 microg/kg, intra-arterially, i.a.) was injected. Two hours later, animals were euthanized, and their small intestines were removed for histological examination. For biochemical studies, PAF (1.5 microg/kg i.a.) was administered and the small intestine removed after 15-60 min. We found that PAF induced an increase in p105 processing within 30 min, but there were no changes in the levels of the NF-kappaB inhibitory proteins IkappaBalpha and beta. NF-kappaB p50-deficient mice were protected against PAF-induced mortality, shock, intestinal hypoperfusion, and injury compared with wild-type animals. We also found that p50-deficient mice had decreased gene expression of CXCL2 and TNF and a decrease in CXCL2 protein production compared with wild-type mice. Our study suggests that PAF increases the processing of NF-kappaB p105 into p50, with upregulation of proinflammatory cytokines, which leads to PAF-induced systemic inflammatory response and acute bowel injury.
Collapse
Affiliation(s)
- Shirley X. L. Liu
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Research Center, Northwestern University Feinberg School of Medicine (Medical School), Chicago, Illinois
| | - Runlan Tian
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Research Center, Northwestern University Feinberg School of Medicine (Medical School), Chicago, Illinois
| | - Heather Baskind
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Research Center, Northwestern University Feinberg School of Medicine (Medical School), Chicago, Illinois
| | - Wei Hsueh
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Research Center, Northwestern University Feinberg School of Medicine (Medical School), Chicago, Illinois
| | - Isabelle G. De Plaen
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Research Center, Northwestern University Feinberg School of Medicine (Medical School), Chicago, Illinois
| |
Collapse
|
15
|
Marotta DM, Costa R, Motta EM, Fernandes ES, Medeiros R, Quintão NL, Campos MM, Calixto JB. Mechanisms underlying the nociceptive responses induced by platelet-activating factor (PAF) in the rat paw. Biochem Pharmacol 2009; 77:1223-35. [DOI: 10.1016/j.bcp.2008.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
16
|
De Plaen IG, Liu SXL, Tian R, Neequaye I, May MJ, Han XB, Hsueh W, Jilling T, Lu J, Caplan MS. Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis. Pediatr Res 2007; 61:716-21. [PMID: 17426653 DOI: 10.1203/pdr.0b013e3180534219] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of morbidity and death in premature infants. NEC is associated with increased levels of pro-inflammatory cytokines in plasma and tissues that are regulated by the transcription factor nuclear factor-kappaB (NF-kappaB). It remains unknown, however, whether NF-kappaB mediates injury in neonatal NEC. We therefore examined the activation status of NF-kappaB perinatally in the small intestine and in a neonatal rat model of NEC. We found that intestinal NF-kappaB is strongly activated at birth and, in dam-fed newborn rats, is down-regulated within a day. In contrast, NF-kappaB remains strongly activated at both d 1 and d 2 in stressed animals, and this is accompanied by a significant decrease in the levels of the endogenous NF-kappaB inhibitor protein IkappaBalpha and IkappaBbeta at d 2. To determine the importance of elevated NF-kappaB activity in intestinal injury in NEC, we administered the NEMO-binding domain (NBD) peptide that selectively inhibits the critical upstream IkappaB kinase (IKK). NBD but not a control peptide decreased mortality and bowel injury in this model, supporting the hypothesis that bowel injury in NEC results from elevated NF-kappaB activity. Our findings therefore lead us to conclude that selective NF-kappaB inhibition represents a promising therapeutic strategy for NEC.
Collapse
Affiliation(s)
- Isabelle G De Plaen
- Department of Pediatrics, Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois 60614, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
De Plaen IG, Han XB, Liu X, Hsueh W, Ghosh S, May MJ. Lipopolysaccharide induces CXCL2/macrophage inflammatory protein-2 gene expression in enterocytes via NF-kappaB activation: independence from endogenous TNF-alpha and platelet-activating factor. Immunology 2006; 118:153-63. [PMID: 16771850 PMCID: PMC1782278 DOI: 10.1111/j.1365-2567.2006.02344.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
CXCL2 (macrophage inflammatory protein-2 (MIP-2)), a critical chemokine for neutrophils, has been shown to be produced in the rat intestine in response to platelet-activating factor (PAF) and to mediate intestinal inflammation and injury. The intestinal epithelium, constantly exposed to bacterial products, is the first line of defence against micro-organisms. It has been reported that enterocytes produce proinflammatory mediators, including tumour necrosis factor (TNF) and PAF, and we showed that lipopolysaccharide (LPS) and TNF activate nuclear factor (NF)-kappaB in enterocytes. However, it remains elusive whether enterocytes release CXCL2 in response to LPS and TNF via a NF-kappaB-dependent pathway and whether this involves the endogenous production of TNF and PAF. In this study, we found that TNF and LPS markedly induced CXCL2 gene expression in IEC-6 cells, TNF within 30 min, peaking at 45 min, while LPS more slowly, peaking after 2 hr. TNF- and LPS- induced CXCL2 gene expression and protein release were completely blocked by pyrrolidine dithiocarbamate (PDTC) and helenalin, two potent NF-kappaB inhibitors. NEMO-binding domain peptide, a specific inhibitor of inhibitor protein kappaB kinase (IKK) activation, a major upstream kinase mediating NF-kappaB activation, significantly blocked CXCL2 gene expression and protein release induced by LPS. WEB2170 (PAF antagonist) and anti-TNF antibodies had no effect on LPS-induced CXCL2 expression. In conclusion, CXCL2 gene is expressed in enterocytes in response to both TNF and LPS. LPS-induced CXCL2 expression is dependent on NF-kappaB activation via the IKK pathway. The effect of LPS is independent of endogenous TNF and PAF.
Collapse
Affiliation(s)
- Isabelle G De Plaen
- Department of Pediatrics (Neonatology), Children's Memorial Hospital, Northwestern University Medical School, Chicago, IL 60614, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Wu J, Nilsson Å, Jönsson B, Stenstad H, Agace W, Cheng Y, Duan RD. Intestinal alkaline sphingomyelinase hydrolyses and inactivates platelet-activating factor by a phospholipase C activity. Biochem J 2006; 394:299-308. [PMID: 16255717 PMCID: PMC1386028 DOI: 10.1042/bj20051121] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Alkaline sphingomyelinase (alk-SMase) is a new member of the NPP (nucleotide pyrophosphatase/phosphodiesterase) family that hydrolyses SM (sphingomyelin) to generate ceramide in the intestinal tract. The enzyme may protect the intestinal mucosa from inflammation and tumorigenesis. PAF (platelet-activating factor) is a pro-inflammatory phospholipid involved in pathogenesis of inflammatory bowel diseases. We examined whether alk-SMase can hydrolyse and inactivate PAF. [3H]Octadecyl-labelled PAF was incubated with purified rat intestinal alk-SMase or recombinant human alk-SMase expressed in COS-7 cells. The hydrolytic products were assayed with TLC and MS. We found that alkSMase cleaved the phosphocholine head group from PAF and generated 1-O-alkyl-2-acetyl-sn-glycerol. Differing from the activity against SM, the activity against PAF was optimal at pH 7.5, inhibited by EDTA and stimulated by 0.1-0.25 mM Zn2+. The activity was abolished by site mutation of the predicted metal-binding sites that are conserved in all NPP members. Similar to the activity against SM, the activity against PAF was dependent on bile salt, particularly taurocholate and taurochenodeoxycholate. The V(max) for PAF hydrolysis was 374 mumol x h(-1) x (mg of protein)(-1). The hydrolysis of PAF and SM could be inhibited by the presence of SM and PAF respectively, the inhibition of PAF hydrolysis by SM being stronger. The PAF-induced MAPK (mitogen-activated protein kinase) activation and IL-8 (interleukin 8) release in HT-29 cells, and chemotaxis in leucocytes were abolished by alk-SMase treatment. In conclusion, alk-SMase hydrolyses and inactivates PAF by a phospholipase C activity. The finding reveals a novel function, by which alk-SMase may counteract the development of intestinal inflammation and colon cancer.
Collapse
Affiliation(s)
- Jun Wu
- *Gastroenterology Laboratory, Biomedical Centre, B11, Lund University, S-221 84 Lund, Sweden
| | - Åke Nilsson
- *Gastroenterology Laboratory, Biomedical Centre, B11, Lund University, S-221 84 Lund, Sweden
| | - Bo A. G. Jönsson
- †Department of Occupational and Environment Medicine, Institute of Laboratory Medicine, University Hospital, S-221 85 Lund, Sweden
| | - Hanna Stenstad
- ‡Immunology Unit, Lund University, S-221 84 Lund, Sweden
| | - William Agace
- ‡Immunology Unit, Lund University, S-221 84 Lund, Sweden
| | - Yajun Cheng
- *Gastroenterology Laboratory, Biomedical Centre, B11, Lund University, S-221 84 Lund, Sweden
| | - Rui-Dong Duan
- *Gastroenterology Laboratory, Biomedical Centre, B11, Lund University, S-221 84 Lund, Sweden
- To whom correspondence should be addressed (email )
| |
Collapse
|
19
|
Colgate EC, Miranda CL, Stevens JF, Bray TM, Ho E. Xanthohumol, a prenylflavonoid derived from hops induces apoptosis and inhibits NF-kappaB activation in prostate epithelial cells. Cancer Lett 2006; 246:201-9. [PMID: 16563612 DOI: 10.1016/j.canlet.2006.02.015] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2005] [Revised: 01/30/2006] [Accepted: 02/20/2006] [Indexed: 11/20/2022]
Abstract
There is increasing evidence that certain natural compounds found in plants may be useful as cancer chemopreventive or chemotherapeutic agents. Limited in vitro studies indicate that several prenylated flavonoids present in the hop plant (Humulus lupulus) possess anticarcinogenic properties. The purpose of this study was to investigate the anti-tumorigenic effects of xanthohumol (XN), the major prenylflavonoid in hops, on prostate cancer and benign prostate hyperplasia. BPH-1 and PC3 cell lines were used in our study to represent both non-tumorigenic hyperplasia and malignant prostate cancer. In both BPH-1 and PC3 cells, XN and its oxidation product, XAL, decreased cell viability in a dose dependent manner (2.5-20 microM) as determined by MTT assay and caused an increase in the formation of early and late apoptotic cells as determined by Annexin V staining and multicaspase assays. XN and its oxygenated derivative also induced cell cycle changes in both cells lines, seen in an elevated sub G1 peak at 48h treatment. Western blot analysis was performed to confirm the activation of proapoptotic proteins, Bax and p53. XN and its derivative caused decreased activation of NFkappaB. This work suggests that XN and its oxidation product, XAL, may be potentially useful as a chemopreventive agent during prostate hyperplasia and prostate carcinogenesis, acting via induction of apoptosis and down-regulation of NFkappaB activation in BPH-1 cells.
Collapse
Affiliation(s)
- Emily C Colgate
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA
| | | | | | | | | |
Collapse
|
20
|
Fernandes ES, Passos GF, Campos MM, de Souza GEP, Fittipaldi JF, Pesquero JL, Teixeira MM, Calixto JB. Cytokines and neutrophils as important mediators of platelet-activating factor-induced kinin B1 receptor expression. Br J Pharmacol 2005; 146:209-16. [PMID: 16025141 PMCID: PMC1576273 DOI: 10.1038/sj.bjp.0706327] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PAF injection into the rat paw is accompanied by the concomitant activation of NF-kappaB and neutrophil influx, which appears to be relevant to the up-regulation of kinin B1 receptors. Herein, we analyse the role of TNF-alpha and IL-1beta production for PAF-induced B1 receptor upregulation in the rat paw. Additionally, we evaluate how cytokine production and neutrophil migration fit into the temporal sequence of events leading to PAF-induced B1 receptor upregulation. In our experiments, treatment with PAF resulted in a marked increase of B1 receptor-mediated paw oedema and in situ production of TNF-alpha at 1 h and IL-1beta at 3 and 6 h later. B1 receptor-mediated paw oedema was significantly inhibited by anti-TNF-alpha antibody and by interleukin-1 receptor antagonist (IRA). TNF-alpha was necessary for the local PAF-induced IL-1beta production. NF-kappaB blocker PDTC prevented the production of both TNF-alpha and IL-1beta, indicating that cytokine production is NF-kappaB dependent. Depletion of neutrophils with an anti-PMN antibody prevented IL-1beta, but not TNF-alpha, production. Although both TNF-alpha and IL-1beta are relevant to functional B1 receptor upregulation, PAF-induced increase in B1 receptor mRNA was markedly suppressed by anti-TNF-alpha and, to a lesser extent, by IRA. B1 receptor mRNA expression was also prevented by the anti-PMN antibody. In conclusion, the activation of the TNF-alpha/neutrophil axis by PAF seems to be sufficient for B1 receptor mRNA production. However, the TNF-alpha/neutrophil axis is also necessary for IL-1beta production. These two processes might lead to the appearance of functional kinin B1 upregulation receptors in vivo after PAF treatment.
Collapse
Affiliation(s)
- Elizabeth S Fernandes
- Department of Pharmacology, CCB, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC 88049-900, Brazil
| | - Giselle F Passos
- Department of Pharmacology, CCB, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC 88049-900, Brazil
| | - Maria M Campos
- Department of Pharmacology, CCB, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC 88049-900, Brazil
| | - Glória E P de Souza
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences of Ribeirão, USP Ribeirão Preto, SP, Brazil
| | | | - Jorge L Pesquero
- Department of Physiology and Biophysics, Belo Horizonte, MG, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - João B Calixto
- Department of Pharmacology, CCB, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC 88049-900, Brazil
- Author for correspondence:
| |
Collapse
|
21
|
Cheikhelard A, Go S, Canioni D, Leborgne M, Brousse N, Révillon Y, Cerf-Bensussan N, Sarnacki S. Enhanced in situ expression of NF-kappaBp65 is an early marker of intestinal graft rejection in rats. J Pediatr Surg 2005; 40:1420-7. [PMID: 16150343 DOI: 10.1016/j.jpedsurg.2005.05.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Although intestinal transplantation provides a unique situation of free access to the graft because of the presence of temporary enterostomas, evaluation of local immunosuppression is still an unresolved issue and may constitute one of the causes of grafting failure. AIMS To study in a rat model of allogeneic intestinal transplantation the expression of transcription factors involved in lymphocyte activation in situ in the graft and to identify factors reflecting the efficiency of drug immunosuppression. METHODS Intestinal transplantation was performed in a Brown Norway (RT1n-donors)-Lewis (RT1(l)-recipients) rat strain combination. The animals were treated with tacrolimus to induce tolerance or left untreated. Syngeneic intestinal grafts and intestine from donor rats with peritonitis were used as controls. NF-kappaBp65, p-c-Jun, interleukin 2 receptor (CD25), and major histocompatibility complex class II antigen (OX-6) expression was studied in graft biopsies on days 2 and 5 by immunohistochemistry. RESULTS On day 2, before the onset of histologic signs of rejection, the number of cells expressing NF-kappaBp65 in the pericryptic lamina propria was significantly higher in untreated recipients of allogeneic grafts than in the other groups (P = .009). NF-kappaBp65 expression then fell between days 2 and 5 (P = .009). Classic markers of T-cell activation (CD25 and OX-6) were expressed during rejection in the lamina propria and on crypt enterocytes, respectively. p-c-Jun expression did not differ among the 3 groups. CONCLUSION NF-kappaBp65 expression in intestinal grafts is a precocious sign of local activation during rejection and could thus serve to optimize the management of immunosuppressive therapy.
Collapse
Affiliation(s)
- Alaa Cheikhelard
- INSERM E-0212, Faculté Necker Enfants-Malades, 75730 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhu YQ, Tan XD. TFF3 modulates NF-{kappa}B and a novel negative regulatory molecule of NF-{kappa}B in intestinal epithelial cells via a mechanism distinct from TNF-{alpha}. Am J Physiol Cell Physiol 2005; 289:C1085-93. [PMID: 16014704 PMCID: PMC2527239 DOI: 10.1152/ajpcell.00185.2005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Trefoil factor 3 (intestinal trefoil factor) is a cytoprotective factor in the gut. Herein we compared the effect of trefoil factor 3 with tumor necrosis factor-alpha on 1) activation of NF-kappaB in intestinal epithelial cells; 2) expression of Twist protein (a molecule essential for downregulation of nuclear factor-kappaB activity in vivo); and 3) production of interleukin-8. We showed that Twist protein is constitutively expressed in intestinal epithelial cells. Tumor necrosis factor-alpha induced persistent degradation of Twist protein in intestinal epithelial cells via a signaling pathway linked to proteasome, which was associated with prolonged activation of NF-kappaB. In contrast to tumor necrosis factor, trefoil factor 3 triggered transient activation of NF-kappaB and prolonged upregulation of Twist protein in intestinal epithelial cells via an ERK kinase-mediated pathway. Unlike tumor necrosis factor-alpha, transient activation of NF-kappaB by trefoil factor 3 is not associated with induction of IL-8 in cells. To examine the role of Twist protein in intestinal epithelial cells, we silenced the Twist expression by siRNA. Our data showed that trefoil factor 3 induced interleukin-8 production after silencing Twist in intestinal epithelial cells. Together, these observations indicated that 1) trefoil factor 3 triggers a diverse signal from tumor necrosis factor-alpha on the activation of NF-kappaB and its associated molecules in intestinal epithelial cells; and 2) trefoil factor 3-induced Twist protein plays an important role in the modulation of inflammatory cytokine production in intestinal epithelial cells.
Collapse
Affiliation(s)
- Ya-Qin Zhu
- Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Chicago, IL 60614
- Departments of Pathology and Pediatrics, Children’s Memorial Hospital, Feinberg School of Medicine, Northwestern University, Chicago, IL 60614
- Ya-Qin Zhu, M.D., functions as the co-corresponding author and could be contacted at
for the correspondence and requesting reprints
| | - Xiao-Di Tan
- Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Chicago, IL 60614
- Departments of Pathology and Pediatrics, Children’s Memorial Hospital, Feinberg School of Medicine, Northwestern University, Chicago, IL 60614
- Address all correspondence to Xiao-Di Tan, M.D. Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Children’s Memorial Hospital, 2300 Children’s Plaza, Box 217, Chicago, IL 60614, Tel: (773) 755-6380, Fax: (773) 755-6581, e-mail:
| |
Collapse
|
23
|
Bai Y, Onuma H, Bai X, Medvedev AV, Misukonis M, Weinberg JB, Cao W, Robidoux J, Floering LM, Daniel KW, Collins S. Persistent nuclear factor-kappa B activation in Ucp2-/- mice leads to enhanced nitric oxide and inflammatory cytokine production. J Biol Chem 2005; 280:19062-9. [PMID: 15757894 PMCID: PMC1382174 DOI: 10.1074/jbc.m500566200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
One of the phenotypes of mice with targeted disruption of the uncoupling protein-2 gene (Ucp2-/-) is greater macrophage phagocytic activity and free radical production, resulting in a striking resistance to infectious microorganisms. In this study, the molecular mechanisms of this enhanced immune response were investigated. We found that levels of nitric oxide measured in either plasma or isolated macrophages from Ucp2-/- mice are significantly elevated in response to bacterial lipopolysaccharide challenge compared with similarly treated Ucp2+/+ mice. Likewise, expression of inducible nitric-oxide synthase and inflammatory cytokines is higher in Ucp2-/- mice in vivo and in vitro. Key steps in the activation cascade of nuclear factor (NF)-kappa B, including I kappa B kinase and nuclear translocation of NF-kappa B subunits, are all remarkably enhanced in Ucp2-/- mice, most notably even under basal conditions. The elevated basal activity of I kappa B kinase in macrophages from Ucp2-/- mice can be blocked by cell-permeable inhibitors of superoxide and hydrogen peroxide generation, but not by a specific inhibitor for inducible nitric-oxide synthase. Isolated mitochondria from Ucp2-/- cells produced more superoxide/hydrogen peroxide. We conclude that mitochrondrially derived reactive oxygen from Ucp2-/- cells constitutively activates NF-kappa B, resulting in a "primed" state to both potentiate and amplify the inflammatory response upon subsequent stimulation.
Collapse
Affiliation(s)
- Yushi Bai
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Hiroki Onuma
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Xu Bai
- Department of Psychiatry and Behavioral Sciences and
| | | | - Mary Misukonis
- Division of Hematology-Oncology, Veterans Administration Medical Center/Duke University Medical Center, Durham, North Carolina 27710
| | - J. Brice Weinberg
- Division of Hematology-Oncology, Veterans Administration Medical Center/Duke University Medical Center, Durham, North Carolina 27710
| | - Wenhong Cao
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Jacques Robidoux
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Lisa M. Floering
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Kiefer W. Daniel
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
| | - Sheila Collins
- From the Division of Biological Sciences, Endocrine Biology Program, CIIT Centers for Health Research, Research Triangle Park, North Carolina 27709-2137 and
- Department of Psychiatry and Behavioral Sciences and
- ||To whom correspondence should be addressed: Endocrine Biology Program, CIIT Centers for Health Research, 6 Davis Dr., P. O. Box 12137, Research Triangle Park, NC 27709-2137. Tel.: 919-558-1378; Fax: 919-558-1305; E-mail:
| |
Collapse
|
24
|
Tong X, Yin L, Washington R, Rosenberg DW, Giardina C. The p50-p50 NF-kappaB complex as a stimulus-specific repressor of gene activation. Mol Cell Biochem 2005; 265:171-83. [PMID: 15543947 DOI: 10.1023/b:mcbi.0000044394.66951.4d] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The transcription factor NF-kappaB can be activated in different forms, including transcriptional activating and repressing forms. Intestinal epithelial cells have been found to modulate the relative levels of the p65-p50 and p50-p50 NF-kappaB complexes in a number of instances, and here we show that this ratio was altered in response to dietary fiber (wheat bran) and carcinogen exposure (azoxymethane). The influence of these complexes on gene regulation was examined in more detail in cell culture models. The colon-derived HT-29 cell line likewise activated both p65-p50 and p50-p50 NF-kappaB complexes: TNF-alpha triggered a strong, sustained p65-p50 activation with lower relative levels of p50-p50, whereas IL-1beta transiently activated p65-p50 with higher relative levels of p50-p50. Transfection experiments with an NF-kappaB reporter plasmid indicated that p50 was a repressor in HT-29 cells. Increased expression of the p50-p50 dimer by an adenovirus showed that the p50-p50 dimer suppressed IL-1beta activation of endogenous genes more than 5-fold (TNF-alpha, Cox-2 and IL-8), whereas gene activation by TNF-alpha was not significantly affected. DNA binding analyses showed a number of strong p50-p50 binding sites on these promoters. The selective p50-p50 suppression of IL-1beta gene activation corresponded to the transient nature of p65-p50 activation induced by IL-1beta (in both HT-29 and Caco-2 cells). Our findings demonstrate a novel gene regulatory mechanism for the NF-kappaB p50-p50 complex: a signal-specific transcriptional repression that appears to selectively inhibit stimuli that transiently activate p65-p50 complexes.
Collapse
Affiliation(s)
- Xin Tong
- Department of Molecular & Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | | | | | | | | |
Collapse
|
25
|
Hang CH, Shi JX, Li JS, Li WQ, Yin HX. Up-regulation of intestinal nuclear factor kappa B and intercellular adhesion molecule-1 following traumatic brain injury in rats. World J Gastroenterol 2005; 11:1149-54. [PMID: 15754395 PMCID: PMC4250704 DOI: 10.3748/wjg.v11.i8.1149] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Nuclear factor kappa B (NF-κB) regulates a large number of genes involved in the inflammatory response to critical illnesses, but it is not known if and how NF-κB is activated and intercellular adhesion molecule-1 (ICAM-1) expressed in the gut following traumatic brain injury (TBI). The aim of current study was to investigate the temporal pattern of intestinal NF-κB activation and ICAM-1 expression following TBI.
METHODS: Male Wistar rats were randomly divided into six groups (6 rats in each group) including controls with sham operation and TBI groups at hours 3, 12, 24, and 72, and on d 7. Parietal brain contusion was adopted using weight-dropping method. All rats were decapitated at corresponding time point and mid-jejunum samples were taken. NF-κB binding activity in jejunal tissue was measured using EMSA. Immunohistochemistry was used for detection of ICAM-1 expression in jejunal samples.
RESULTS: There was a very low NF-κB binding activity and little ICAM-1 expression in the gut of control rats after sham surgery. NF-κB binding activity in jejunum significantly increased by 160% at 3 h following TBI (P<0.05 vs control), peaked at 72 h (500% increase) and remained elevated on d 7 post-injury by 390% increase. Compared to controls, ICAM-1 was significantly up-regulated on the endothelia of microvessels in villous interstitium and lamina propria by 24 h following TBI and maximally expressed at 72 h post-injury (P<0.001). The endothelial ICAM-1 immunoreactivity in jejunal mucosa still remained strong on d 7 post-injury. The peak of NF-κB activation and endothelial ICAM-1 expression coincided in time with the period during which secondary mucosal injury of the gut was also at their culmination following TBI.
CONCLUSION: TBI could induce an immediate and persistent up-regulation of NF-κB activity and subsequent up-regulation of ICAM-1 expression in the intestine. Inflammatory response mediated by increased NF-κB activation and ICAM-1 expression may play an important role in the pathogenesis of acute gut mucosal injury following TBI.
Collapse
Affiliation(s)
- Chun-Hua Hang
- Department of Neurosurgery, Jinling Hospital, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, China.
| | | | | | | | | |
Collapse
|
26
|
Han XB, Liu X, Hsueh W, De Plaen IG. Macrophage inflammatory protein-2 mediates the bowel injury induced by platelet-activating factor. Am J Physiol Gastrointest Liver Physiol 2004; 287:G1220-6. [PMID: 15319184 DOI: 10.1152/ajpgi.00231.2004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Platelet-activating factor (PAF) is a potent endogenous mediator of bowel inflammation. It activates neutrophils that are needed to initiate the inflammatory response. Macrophage inflammatory protein-2 (MIP-2), a critical C-X-C chemokine secreted by macrophages and epithelial cells, is a potent chemoattractant for neutrophils. Whereas MIP-2 has been previously shown to mediate the injury in various organs, its role in acute intestinal injury has never been assessed. In this study, we first investigated the effect of PAF on MIP-2 expression in the intestine. Anesthetized young adult male Sprague-Dawley rats were injected intravenously with either PAF (1.5 microg/kg) or saline. Sixty minutes later, ileal MIP-2 gene expression was determined by semiquantitative RT-PCR, and plasma and ileal MIP-2 protein was determined by ELISA. In a second step, we assessed the role of MIP-2 in PAF-induced bowel injury. Rats were pretreated with rabbit anti-rat MIP-2 antibodies or control IgG for 90 min and then injected intravenously with PAF (2.5 microg/kg) for 90 min. We found that, in the rat intestine, 1) MIP-2 mRNA was only minimally expressed constitutively in sham-operated animals; 2) MIP-2 mRNA was significantly upregulated in response to PAF; 3) MIP-2 protein plasma levels and local production of MIP-2 in the ileum were markedly induced by PAF; 4) the administration of anti-rat MIP-2 IgG, but not control rabbit IgG, markedly reduced PAF-induced bowel injury (injury scores of 0.19 +/- 0.09 vs. 1.12 +/- 0.43, P < 0.05), hypotension, and leukopenia but did not reduce PAF-induced hemoconcentration. Thus we conclude that MIP-2 mediates PAF-induced intestinal injury.
Collapse
Affiliation(s)
- Xin-Bing Han
- Department of Pediatrics, Children's Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL 60614, USA
| | | | | | | |
Collapse
|
27
|
Minekawa R, Takeda T, Sakata M, Hayashi M, Isobe A, Yamamoto T, Tasaka K, Murata Y. Human breast milk suppresses the transcriptional regulation of IL-1beta-induced NF-kappaB signaling in human intestinal cells. Am J Physiol Cell Physiol 2004; 287:C1404-11. [PMID: 15229109 DOI: 10.1152/ajpcell.00471.2003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neonatal necrotizing enterocolitis (NEC), which is a disease with a poor prognosis, is considered to be caused by the coincidence of intestinal ischemia-reperfusion injury and systemic inflammation due to the colonization of pathogenic bacteria. Interleukin (IL)-8, a proinflammatory cytokine, plays an important role in the pathophysiology of NEC. It was recently reported that IL-1beta activates the IL-8 gene by regulating the transcriptional nuclear factor kappaB (NF-kappaB) signaling pathways in intestinal cells. The protective role of maternal milk in NEC pathogenesis has been reported in both human and animal studies. In this study, we show that human breast milk dramatically suppressed the IL-1beta-induced activation of the IL-8 gene promoter by inhibiting the activation pathway of NF-kappaB. Moreover, we also show that human breast milk induced the production of IkappaBalpha. These results suggest that human breast milk could be protective and therapeutic in neonates with NEC by inhibiting the activation pathway of NF-kappaB.
Collapse
Affiliation(s)
- Ryoko Minekawa
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Seyffarth G, Nelson PN, Dunmore SJ, Rodrigo N, Murphy DJ, Carson RJ. Lipopolysaccharide induces nitric oxide synthase expression and platelet-activating factor increases nitric oxide production in human fetal membranes in culture. Reprod Biol Endocrinol 2004; 2:29. [PMID: 15191613 PMCID: PMC434531 DOI: 10.1186/1477-7827-2-29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2004] [Accepted: 06/10/2004] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Platelet-activating factor and nitric oxide may be involved in the initiation of human labour as inflammatory mediators. The aim of this study was to test whether platelet-activating factor and lipopolysaccharide were able to induce nitric oxide synthase expression and stimulate the production of nitric oxide in human fetal membrane explants in culture. METHODS Fetal membranes were collected from Caesarean sections at term. RNA was extracted from membranes and subjected to a qualitative RT-PCR to assess the baseline expression of iNOS. Discs of fetal membranes were cultured for 24 hours in the presence of platelet-activating factor at a dose range of 0.1 nanomolar--1 micomolar or 1 microgram/ml lipopolysaccharide. Nitric oxide production was measured via nitrite ions in the culture medium and mRNA for iNOS was detected by RT-PCR. RESULTS Culturing the membrane discs in medium containing serum induced nitric oxide synthase expression and platelet-activating factor significantly stimulated the production of nitric oxide under these conditions. When cultured without serum inducible nitric oxide synthase expression was induced by lipopolysaccharide, but not by platelet-activating factor. CONCLUSION Platelet-activating factor may have a role in the initiation of labour, at term or preterm, via the increased local production of nitric oxide as an inflammatory mediator. In this model of intrauterine infection, lipopolysaccharide was found to induce iNOS expression by fetal membranes, and this mechanism could be involved in preterm labour.
Collapse
Affiliation(s)
- Gunter Seyffarth
- Perinatal and Maternal Studies Group, University of Wolverhampton, UK
| | - Paul N Nelson
- Molecular Immunology Research Group, Division of Biomedical Sciences, University of Wolverhampton, UK
| | - Simon J Dunmore
- Diabetes Group, School of Applied Science, University of Wolverhampton, UK
| | | | | | - Ray J Carson
- Physiology Section, School of Science and the Environment, Coventry University, Priory Street, Coventry, CV1 5FB, UK
| |
Collapse
|
29
|
Cerezo CS, Kulpa-Oliver V, Gruppuso PA, Morin MJ. REGULATION OF ADOLESCENT RAT INTESTINAL EPITHELIAL INDUCIBLE NITRIC OXIDE SYNTHASE EXPRESSION IN ENDOTOXIN TOLERANCE: MODULATION OF SIGNAL TRANSDUCTION. Shock 2004; 21:476-83. [PMID: 15087826 DOI: 10.1097/00024382-200405000-00013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Endotoxin/lipopolysaccharide (LPS) tolerance is a state of induced hyporesponsiveness to endotoxin or LPS characterized by alterations in the release of inflammatory mediators. As the gut is both a source of infection and target of injury, we tested the hypothesis that alterations in intestinal epithelial signal transduction would account for the acquisition of endotoxin tolerance as defined by decreased induction of a key mediator of gut injury, inducible nitric oxide synthase (iNOS). Rats (15 days of age) were injected with saline or LPS (1 microg/g i.p.). Tissue was harvested after 1, 4, or 6 h for assessment of signaling and iNOS expression. Other animals received a second dose of LPS 1 to 7 days after the initial dose. Selected animals received the p38 inhibitor, SB203580 (10 microg/g), which was co-administered with the first dose of LPS. Induction of iNOS mRNA and protein was significantly attenuated after repeated LPS administration. Epithelial cells from LPS-tolerant rats showed a minimal level of iNOS expression by immunohistochemistry. The down-regulation of intestinal iNOS was not gender dependent. p38 inhibition enhanced tolerance rather than blocking it. LPS-mediated activation of NF-kappaB was attenuated in a manner consistent with a primary role in the induction of tolerance. Endotoxin tolerance can be demonstrated in intestinal epithelial cells using an in vivo model. Modulation of NF-kappaB signaling may be key in the down-regulation of LPS effect seen in tolerance.
Collapse
Affiliation(s)
- Caroline S Cerezo
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, Rhode Island 02903, USA
| | | | | | | |
Collapse
|
30
|
Fernandes ES, Passos GF, Campos MM, Araújo JGVC, Pesquero JL, Avelllar MC, Teixeira MM, Calixto JB. Mechanisms underlying the modulatory action of platelet activating factor (PAF) on the upregulation of kinin B1 receptors in the rat paw. Br J Pharmacol 2003; 139:973-81. [PMID: 12839871 PMCID: PMC1573912 DOI: 10.1038/sj.bjp.0705314] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The present study evaluated the ability of the administration of platelet activating factor (PAF) to induce the upregulation of B(1) receptors in the rat paw. 2. Local treatment with PAF resulted in a time-dependent increase of oedema formation induced by the B(1) receptor agonist des-Arg(9)-BK (des-Arg(9)-bradykinin), but not by the B(2) receptor agonist tyrosine(8)-bradykinin. Functional upregulation of B(1) receptors was accompanied by a prominent increase of B(1) receptor mRNA expression in the rat paw. 3. In PAF-treated paws, des-Arg(9)-BK-induced oedema formation was significantly inhibited by the B(1) receptor antagonists des-Arg(9)-[Leu(8)]-BK and R-715. The effects of PAF pretreatment were receptor operated, as assessed by the effects of the PAF receptor antagonist WEB2086 or by desensitisation of PAF receptors. 4. The protein synthesis inhibitor cycloheximide, the anti-inflammatory steroid dexamethasone or the nuclear factor (NF-kappaB) blockers pyrrolidine-dithiocarbamate (PDTC) and Nalpha-tosyl-L-chloromethylketone significantly blocked the functional upregulation of B(1) receptors. 5. The selectin inhibitor fucoidin, an anti-CD18 antibody or an anti-rat neutrophil antiserum, also significantly prevented des-Arg(9)-BK-induced paw oedema in rats pretreated with PAF. 6. Intradermal injection of PAF induced a 25-fold increase of myeloperoxidase activity in the rat paw, a response that was significantly inhibited by fucoidin, anti-CD-18, anti-rat neutrophil antiserum or PDTC. 7. Local treatment with PAF also resulted in a marked increase of NF-kappaB activation, an effect largely prevented by PDTC or by the anti-rat neutrophil antiserum. 8. Collectively, the present results indicate that the induction of B(1) receptors following treatment with the chemotatic mediator PAF is dependent on the recruitment of neutrophils, an event that is under the control of adhesion molecules, protein synthesis and NF-kappaB activation. These findings provide new insights into the role played by cell migration and chemotatic factors on B(1) receptor upregulation in vivo.
Collapse
Affiliation(s)
- Elizabeth S Fernandes
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Rua Ferreira Lima, 82, Florianópolis, SC 88015-420, Brazil
| | - Giselle F Passos
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Rua Ferreira Lima, 82, Florianópolis, SC 88015-420, Brazil
| | - Maria M Campos
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Rua Ferreira Lima, 82, Florianópolis, SC 88015-420, Brazil
| | - José G V C Araújo
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jorge L Pesquero
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria C Avelllar
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo – Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - João B Calixto
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Rua Ferreira Lima, 82, Florianópolis, SC 88015-420, Brazil
- Author for correspondence:
| |
Collapse
|
31
|
Hsueh W, Caplan MS, Qu XW, Tan XD, De Plaen IG, Gonzalez-Crussi F. Neonatal necrotizing enterocolitis: clinical considerations and pathogenetic concepts. Pediatr Dev Pathol 2003; 6:6-23. [PMID: 12424605 PMCID: PMC7098425 DOI: 10.1007/s10024-002-0602-z] [Citation(s) in RCA: 275] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2002] [Accepted: 08/21/2002] [Indexed: 12/13/2022]
Abstract
Necrotizing enterocolitis (NEC), a disease affecting predominantly premature infants, is a leading cause of morbidity and mortality in neonatal intensive care units. Although several predisposing factors have been identified, such as prematurity, enteral feeding, and infection, its pathogenesis remains elusive. In the past 20 years, we have established several animal models of NEC in rats and found several endogenous mediators, especially platelet-activating factor (PAF), which may play a pivotal role in NEC. Injection of PAF induces intestinal necrosis, and PAF antagonists prevent the bowel injury induced by bacterial endotoxin, hypoxia, or challenge with tumor necrosis factor-a (TNF) plus endotoxin in adult rats. The same is true for lesions induced by hypoxia and enteral feeding in neonatal animals. Human patients with NEC show high levels of PAF and decreased plasma PAF-acetylhydrolase, the enzyme degrading PAF. The initial event in our experimental models of NEC is probably polymorphonuclear leukocyte (PMN) activation and adhesion to venules in the intestine, which initiates a local inflammatory reaction involving proinflammatory mediators including TNF, complement, prostaglandins, and leukotriene C4. Subsequent norepinephrine release and mesenteric vasoconstriction result in splanchnic ischemia and reperfusion. Bacterial products (e.g., endotoxin) enter the intestinal tissue during local mucosal barrier breakdown, and endotoxin synergizes with PAF to amplify the inflammation. Reactive oxygen species produced by the activated leukocytes and by intestinal epithelial xanthine oxidase may be the final pathway for tissue injury. Protective mechanisms include nitric oxide produced by the constitutive (mainly neuronal) nitric oxide synthase, and indigenous probiotics such as Bifidobacteria infantis. The former maintains intestinal perfusion and the integrity of the mucosal barrier, and the latter keep virulent bacteria in check. The development of tissue injury depends on the balance between injurious and protective mechanisms.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Disease Models, Animal
- Enterocolitis, Necrotizing/blood
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/pathology
- Humans
- Infant, Newborn
- Infant, Newborn, Diseases/blood
- Infant, Newborn, Diseases/etiology
- Infant, Newborn, Diseases/pathology
- Platelet Activating Factor/analysis
- Species Specificity
Collapse
Affiliation(s)
- Wei Hsueh
- Department of Pathology, Children's Memorial Hospital, Northwestern University Medical School, 2300 Children's Plaza, Chicago, IL 60614, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Li J, Quan N, Bray TM. Supplementation of N-acetylcysteine normalizes lipopolysaccharide-induced nuclear factor kappaB activation and proinflammatory cytokine production during early rehabilitation of protein malnourished mice. J Nutr 2002; 132:3286-92. [PMID: 12421841 DOI: 10.1093/jn/132.11.3286] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Increased sensitivity to septic shock has been reported in protein malnourished patients. In this study, we used an animal septic shock model to investigate effects of glutathione (GSH) levels on nuclear factor kappaB (NFkappaB) activation and proinflammatory cytokine production in protein malnutrition. We further investigated molecular mechanisms by which protein malnutrition influenced inflammatory responses. CD-1 mice were fed for 3 wk a normal protein (150 g/kg) diet or a protein-deficient (5 g/kg) diet, or for 2 wk a protein-deficient diet followed by 1 wk of N-acetylcysteine (NAC) supplementation. Lipopolysaccharide (LPS) was injected intravenously, and liver was collected at 0, 15 min, 1, 4, 24 and 48 h after LPS administration. Protein malnutrition significantly increased the activation of NFkappaB and transcription levels of its downstream genes interleukin-1beta and tumor necrosis factor-alpha. Peak NFkappaB activation was inversely associated with GSH levels (r = -0.939, P < 0.0001) but positively correlated with the GSH disulfide/2GSH reduction potential (r = 0.944 P < 0.0001). We noted unusual NFkappaB p50/p50 homodimer translocation that was significantly elevated in tissue from protein malnourished mice, along with decreased peak levels of normal p65/p50 heterodimer translocation. Interestingly, mRNA levels of IkappaB-alpha were not affected by protein malnutrition. However, early supplementation of NAC to protein malnourished mice without replenishing with dietary protein restored GSH levels and reduction potential, and normalized NFkappaB activation and proinflammatory cytokine production. Taken together, these findings provide evidence supporting the role of GSH in NFkappaB activation and inflammatory response in protein malnutrition, and the use of NAC in early rehabilitation of protein malnutrition without a high protein diet.
Collapse
Affiliation(s)
- Jun Li
- Department of Human Nutrition, School of Dentistry, Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
33
|
De Plaen IG, Qu XW, Wang H, Tan XD, Wang L, Han XB, Rozenfeld RA, Hsueh W. Endotoxin, but not platelet-activating factor, activates nuclear factor-kappaB and increases IkappaBalpha and IkappaBbeta turnover in enterocytes. Immunology 2002; 106:577-83. [PMID: 12153521 PMCID: PMC1782741 DOI: 10.1046/j.1365-2567.2002.01453.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial endotoxin (lipopolysaccharide; LPS) and platelet-activating factor (PAF) are important triggers of bowel inflammation and injury. We have previously shown that LPS activates the transcription factor nuclear factor (NF)-kappaB in the intestine, which up-regulates many pro-inflammatory genes. This effect partly depends on neutrophils and endogenous PAF. However, whether LPS and PAF directly activate NF-kappaB in enterocytes remains controversial. In this study, we first investigated the effect of LPS and PAF on NF-kappaB activation in IEC-6 (a non-transformed rat small intestinal crypt cell line) cells, by electrophoresis mobility shift assay and supershift, and found that LPS, but not PAF, activates NF-kappaB mostly as p50-p65 heterodimers. The effect was slower than tumour necrosis factor (TNF). Both LPS and TNF induce the expression of the NF-kappaB-dependent gene inducible nitric oxide synthase (iNOS), which occurs subsequent to NF-kappaB activation. We then examined the effect of LPS and TNF on the inhibitory molecules IkappaBalpha and IkappaBbeta. We found that TNF causes rapid degradation of IkappaBalpha and IkappaBbeta. In contrast, LPS did not change the levels of IkappaBalpha and IkappaBbeta up to 4 hr (by Western blot). However, in the presence of cycloheximide, there was a slow reduction of IkappaBalpha and IkappaBbeta, which disappeared almost completely at 4 hr. These observations suggest that LPS causes slow degradation and synthesis of IkappaBalpha and IkappaBbeta and therefore activates NF-kappaBeta via at least two mechanisms: initially, through an IkappaB-independent mechanism, and later, via an increased turnover of the inhibitor IkappaB. NF-kappaBeta activation precedes the gene expression of iNOS (assayed by reverse transcription-polymerase chain reaction), suggesting that LPS up-regulates iNOS via this transcription factor.
Collapse
Affiliation(s)
- Isabelle G De Plaen
- Departments of Pediatrics (Neonatology) and Pathology, Children's Memorial Hospital, Northwestern University Medical School, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Hassoun HT, Zou L, Moore FA, Kozar RA, Weisbrodt NW, Kone BC. Alpha-melanocyte-stimulating hormone protects against mesenteric ischemia-reperfusion injury. Am J Physiol Gastrointest Liver Physiol 2002; 282:G1059-68. [PMID: 12016132 DOI: 10.1152/ajpgi.00073.2001] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mesenteric ischemia-reperfusion (I/R) injury to the intestine is a common and often devastating clinical occurrence for which there are few therapeutic options. alpha-Melanocyte-stimulating hormone (alpha-MSH) is a tridecapeptide released by the pituitary gland and immunocompetent cells that exerts anti-inflammatory actions and abrogates postischemic injury to the kidneys and brainstem of rodents. To test the hypothesis that alpha-MSH would afford similar protection in the postischemic small intestine, we analyzed the effects of this peptide on intestinal transit, histology, myeloperoxidase activity, and nuclear factor-kappaB (NF-kappaB) activation after 45 min of superior mesenteric artery occlusion and <or=6 h of reperfusion. Rats subjected to I/R exhibited markedly depressed intestinal transit, histological evidence of severe injury to the ileum, increased myeloperoxidase activity in ileal cytoplasmic extracts, and biphasic activation of NF-kappaB in ileal nuclear extracts. In contrast, rats treated with alpha-MSH before I/R exhibited intestinal transit and histological injury scores comparable to those of sham-operated controls. In addition, the alpha-MSH-treated rats demonstrated less I/R-induced activation of intestinal NF-kappaB and myeloperoxidase activity after prolonged (6 h) reperfusion. We conclude that alpha-MSH significantly limits postischemic injury to the rat small intestine.
Collapse
Affiliation(s)
- Heitham T Hassoun
- Department of Surgery, University of Texas Medical School at Houston, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
35
|
Pritts T, Hungness E, Wang Q, Robb B, Hershko D, Hasselgren PO. Mucosal and enterocyte IL-6 production during sepsis and endotoxemia--role of transcription factors and regulation by the stress response. Am J Surg 2002; 183:372-83. [PMID: 11975924 DOI: 10.1016/s0002-9610(02)00812-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Sepsis and endotoxemia are associated with increased production of interleukin-6 (IL-6) in gut mucosa. Mucosal IL-6 may regulate enterocyte acute phase protein synthesis and intestinal IgA production. In addition, increased IL-6 has been proposed to be a mechanism of loss of mucosal integrity in critical illness. The purpose of this review is to describe current knowledge of the regulation of IL-6 production in the enterocyte/mucosa during inflammation caused by sepsis and endotoxemia. DATA SOURCES Recent publications describing the influence of sepsis, endotoxemia, and proinflammatory cytokines on mucosal/enterocyte IL-6 production. CONCLUSIONS IL-6 production is increased in gut mucosa during sepsis and endotoxemia and in cultured enterocytes after treatment with endotoxin or proinflammatory cytokines. The IL-6 gene is regulated by multiple transcription factors, including NF-kappaB, AP-1, and C/EBP. Because of the multiple important biological roles of IL-6, understanding the cellular and molecular mechanisms of mucosal/enterocyte IL-6 production as well as methods to modulate IL-6 production is of clinical importance in the setting of sepsis and other critical illness.
Collapse
Affiliation(s)
- Timothy Pritts
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | | | | | | | | | | |
Collapse
|
36
|
Wang H, Qu X, De Plaen IG, Hsueh W. Platelet-activating factor and endotoxin activate CCAAT/enhancer binding protein in rat small intestine. Br J Pharmacol 2001; 133:713-21. [PMID: 11429396 PMCID: PMC1572822 DOI: 10.1038/sj.bjp.0704102] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The transcription factor family CCAAT/enhancer binding proteins (C/EBP) is involved in inflammation via the regulation of the gene expression of various pro-inflammatory cytokines and proteins. PAF and endotoxin (lipopolysaccharide, LPS) are known agents causing intestinal inflammation and injury. In this study, we examined the binding activity of C/EBP isoforms in rat small intestine in response to PAF (1.5 microg kg(-1), i.v.) or LPS (5 mg kg(-1), i.v.). We found that C/EBP is constitutively active in normal small intestine, mainly as C/EBP-alpha and beta (C/EBP-beta>alpha). Both C/EBP-alpha and beta are localized in the intestinal epithelial cells: C/EBP-alpha mainly in the crypts, and C/EBP-beta in both villi and crypts, as well as in some lamina propria cells. Only minute amounts of C/EBP-delta were found. PAF rapidly upregulates the binding activity of C/EBP-alpha and beta within 30 min. The increase in C/EBP-alpha is prominent in the crypt cells, whereas the change of C/EBP-beta is more widespread. LPS also increases the binding activity of C/EBP-alpha and beta, and the response is slower than PAF. PAF synergizes with LPS to markedly activate all three subunits. The increase in C/EBP-alpha is transient, whereas the other two have a sustained elevation until 120 min. After challenge with PAF (but not LPS), small amounts of nuclear factor -kappaB (NF-kappaB) p50 and p65 subunits are found in the C/EBP-DNA binding complex, indicating cross-dimerization of the two transcription families. Pretreatment of rats with pyrrolidine dithiocarbamate (PDTC) suppresses LPS-, but not PAF-, induced NF-kappaB and C/EBP binding activity, and significantly increases the C/EBP-delta subunit in LPS- or PAF-induced C/EBP complex. These results suggest that PAF and LPS activate intestinal C/EBP in vivo, probably via different pathways.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pathology, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Xiaowu Qu
- Department of Pathology, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Isabelle G De Plaen
- Department of Pediatrics, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Wei Hsueh
- Department of Pathology, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
- Author for correspondence:
| |
Collapse
|
37
|
Hummel M, Zhang Z, Yan S, DePlaen I, Golia P, Varghese T, Thomas G, Abecassis MI. Allogeneic transplantation induces expression of cytomegalovirus immediate-early genes in vivo: a model for reactivation from latency. J Virol 2001; 75:4814-22. [PMID: 11312353 PMCID: PMC114236 DOI: 10.1128/jvi.75.10.4814-4822.2001] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Reactivation of cytomegalovirus (CMV) from latency is a frequent complication of organ transplantation, and the molecular mechanism by which this occurs is unknown. Previous studies have shown that allogeneic stimulation induces reactivation of human CMV (HCMV) in vitro (64). We find that transplantation of vascularized allogeneic kidneys induces murine CMV (MCMV) and HCMV immediate-early (ie) gene expression. This induction is accompanied by increased expression of transcripts encoding inflammatory cytokines, including tumor necrosis factor (TNF), interleukin-2, and gamma interferon, and by activation of NF-kappaB. TNF alone can substitute for allogeneic transplantation in inducing HCMV and MCMV ie gene expression in some tissues. Our studies suggest that reactivation is a multistep process which is initiated by factors that induce ie gene expression, including TNF and NF-kappaB. Allogeneic transplantation combined with immunosuppression may be required to achieve complete reactivation in vivo.
Collapse
Affiliation(s)
- M Hummel
- Department of Surgery, Division of Organ Transplantation, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Qu XW, Wang H, De Plaen IG, Rozenfeld RA, Hsueh W. Neuronal nitric oxide synthase (NOS) regulates the expression of inducible NOS in rat small intestine via modulation of nuclear factor kappa B. FASEB J 2001; 15:439-46. [PMID: 11156959 DOI: 10.1096/fj.99-0343com] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We previously reported that neuronal nitric oxide synthase (nNOS) is the predominant NOS in the intestine. Inducible NOS (iNOS), an enzyme involved in the inflammatory response, is regulated by cytokines via the transcriptional factor NF-kappaB. We examined a new mechanism of intestinal iNOS regulation with respect to the role of nNOS and its effect on NF-kappaB. Young Sprague-Dawley rats were treated for 4 days with 1) saline, 2) 7-nitroindazole (7-NI, specific nNOS inhibitor), 3) 7-NI + pyrrolidine dithiocarbamate (PDTC, NF-kappaB inhibitor), or 4) PDTC. Intestinal iNOS mRNA, NF-kappaB activity, and the tissue content of the regulatory IkappaBalpha were examined. We found that 7-NI-treated animals had higher intestinal NF-kappaB (p50-p65) activity, lower IkappaBalpha content, and increased intestinal iNOS mRNA, iNOS protein, and iNOS activity compared with controls. All of these changes were abolished when PDTC was given together with 7-NI. PDTC alone had no effect. 7-NI induces a delayed increase in intestinal myeloperoxidase activity (after elevation in NF-kappaB and iNOS), which could be abrogated by PDTC. We conclude that in normal rat small intestine, nNOS suppresses the gene expression of iNOS through NF-kappaB down-regulation and that nNOS suppression leads to IkappaBalpha degradation, NF-kappaB activation, and iNOS expression.
Collapse
Affiliation(s)
- X W Qu
- Department of Pathology, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois 60614, USA
| | | | | | | | | |
Collapse
|
39
|
Inan MS, Tolmacheva V, Wang QS, Rosenberg DW, Giardina C. Transcription factor NF-kappaB participates in regulation of epithelial cell turnover in the colon. Am J Physiol Gastrointest Liver Physiol 2000; 279:G1282-91. [PMID: 11093952 DOI: 10.1152/ajpgi.2000.279.6.g1282] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The transcription factor nuclear factor (NF)-kappaB regulates the expression of genes that can influence cell proliferation and death. Here we analyze the contribution of NF-kappaB to the regulation of epithelial cell turnover in the colon. Immunohistochemical, immunoblot, and DNA binding analyses indicate that NF-kappaB complexes change as colonocytes mature: p65-p50 complexes predominate in proliferating epithelial cells of the colon, whereas the p50-p50 dimer is prevalent in mature epithelial cells. NF-kappaB1 (p50) knockout mice were used to study the role of NF-kappaB in regulating epithelial cell turnover. Knockout animals lacked detectable NF-kappaB DNA binding activity in isolated epithelial cells and had significantly longer crypts with a more extensive proliferative zone than their wild-type counterparts (as determined by proliferating cell nuclear antigen staining and in vivo bromodeoxyuridine labeling). Gene expression profiling reveals that the NF-kappaB1 knockout mice express the potentially growth-enhancing tumor necrosis factor (TNF)-alpha and nerve growth factor-alpha genes at elevated levels, with in situ hybridization localizing some of the TNF-alpha expression to epithelial cells. TNF-alpha is NF-kappaB regulated, and its upregulation in NF-kappaB1 knockouts may result from an alleviation of p50-p50 repression. NF-kappaB complexes may therefore influence cell proliferation in the colon through their ability to selectively activate and/or repress gene expression.
Collapse
Affiliation(s)
- M S Inan
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA
| | | | | | | | | |
Collapse
|
40
|
Chen YH, Lu Y, De Plaen IG, Wang LY, Tan XD. Transcription factor NF-kappaB signals antianoikic function of trefoil factor 3 on intestinal epithelial cells. Biochem Biophys Res Commun 2000; 274:576-82. [PMID: 10924320 DOI: 10.1006/bbrc.2000.3176] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transcription factor NF-kappaB has both pro-apoptotic and anti-apoptotic properties depending on the cell type. Its role in the intestinal epithelial cell has not been well elucidated. Trefoil factor 3 (TFF3) is an anti-apoptotic peptide secreted by intestinal goblet cells. Here we show that TFF3 activated NF-kappaB p50/p65 heterodimer within 1 h in IEC-18 cells (a nontransformed rat intestinal epithelial cell line). Moreover, we found that TFF3-treated IEC-18 cells are resistant to anoikis, an anchorage-related apoptosis in epithelium. In addition, the stable expression of a mutant form of the endogenous NF-kappaB inhibitor (IkappaBalpha(mut)) in IEC-18 cells results in a significant attenuation of anti-anoikic effect of TFF3. Taken together, these data indicate that (1) TFF3 is an endogenous gastrointestinal peptide with anti-anoikic property; (2) TFF3 activates NF-kappaB in enterocytes; and (3) TFF3-induced resistance to anoikis in intestinal epithelial cells is mediated by a distinct signaling cascade linked to NF-kappaB. Furthermore, our study implicates NF-kappaB as an important regulator in survival pathway of intestinal epithelial cells.
Collapse
Affiliation(s)
- Y H Chen
- Department of Pathology, Northwestern University, Chicago, Illinois, 60614, USA
| | | | | | | | | |
Collapse
|
41
|
Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C. The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology 2000; 118:724-34. [PMID: 10734024 DOI: 10.1016/s0016-5085(00)70142-9] [Citation(s) in RCA: 338] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The transcription factor nuclear factor-kappaB (NF-kappaB) plays a central role in regulating immune and inflammatory responses. Because butyrate deficiency has been associated with inflammatory bowel disease, we examined the effect of butyrate on NF-kappaB activity in the human HT-29 colonic cell line. METHODS The influence of butyrate (4 mmol/L) on NF-kappaB activity was determined using the gel mobility shift assay. The effect of butyrate on the expression of NF-kappaB subunits and inhibitory proteins was determined by immunoblotting. NF-kappaB-regulated gene expression was assayed by primer extension of intercellular adhesion molecule 1 and Mn superoxide dismutase messenger RNA, and by analysis of a transfected luciferase reporter. RESULTS Exposure of HT-29 cells to butyrate eliminated their constitutive NF-kappaB, p50 dimer activity. This inhibition corresponded with a reduction in p50 nuclear localization, without a reduction in expression. Butyrate also selectively modulated activation of NF-kappaB, suppressing its activation by tumor necrosis factor alpha and phorbol ester more than 10-fold, without affecting the activity induced by interleukin (IL)-1beta. Butyrate did, however, enhance formation of the stronger p65-p50 transcriptional activator in IL-1beta-stimulated cells. The changes in NF-kappaB activation did not correlate with changes in IkappaBalpha levels. Gene expression reflected DNA binding. The influence of butyrate on NF-kappaB may result in part from its ability to inhibit deacetylases because the specific deacetylase inhibitor trichostatin A has a similar effect. CONCLUSIONS These findings suggest that the influences of butyrate on colonic inflammatory responses may result in part from its influence on NF-kappaB activation. This activity of butyrate apparently involves its ability to inhibit deacetylases.
Collapse
Affiliation(s)
- M S Inan
- Department of Molecular Biology, University of Connecticut, Storrs, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
42
|
Tan XD, Chang H, Qu XW, Caplan M, Gonzalez-Crussi F, Hsueh W. Platelet-activating factor increases mucosal permeability in rat intestine via tyrosine phosphorylation of E-cadherin. Br J Pharmacol 2000; 129:1522-9. [PMID: 10742310 PMCID: PMC1571959 DOI: 10.1038/sj.bjp.0702939] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
1. Platelet-activating factor (PAF), an inflammatory mediator, plays an important role in mediating intestinal injury. However, it remains unclear whether PAF has a function in the intestine. The production of PAF by normal intestine and by unstimulated intestinal epithelial cell lines suggests that PAF may have a regulatory function in the normal bowel. 2. In this study we investigated the role of PAF in modulating intestinal mucosal permeability in rats. Lumen-to-blood transit of FD-4 (dextran 4400), (an index of intestinal permeability), was assessed in sham-operated rats and rats injected with PAF (1.25 microg kg(-1), i.v., a dose insufficient to induce intestinal injury). 3. PAF-induced villus cytoskeletal changes were examined by staining the intestine for F-actin. The effect of PAF on tyrosine phosphorylation of the junctional protein E-cadherin was examined by immunoprecipitation. Some rats were pretreated with AG1288 (a tyrosine kinase inhibitor) before PAF injection, and mucosal permeability change was assessed. 4. To investigate the role of endogenous PAF upon mucosal permeability, we studied the effect of PAF antagonists on (intraluminal) glucose-induced increase in mucosal permeability. 5. We found that low dose PAF: (a) alters the cytoskeletal structure of intestinal epithelium, (b) causes the influx of FD4 from intestinal lumen to systemic circulation, (c) induces tyrosine phosphorylation of E-cadherin and cadherin-associated proteins. Glucose-induced mucosal permeability increase is abolished by using two structurally different PAF antagonists. 6. These results suggest that endogenous PAF modulates macromolecular movement across the intestinal mucosal barrier, probably via tyrosine phosphorylation of E-cadherin and cytoskeletal alteration of enterocytes.
Collapse
Affiliation(s)
- Xiao-Di Tan
- Department of Pathology, Children's Memorial Hospital, 2300 Childrens's Plaza, Chicago, IL 60614, U.S.A
| | - Hong Chang
- Department of Pathology, Children's Memorial Hospital, 2300 Childrens's Plaza, Chicago, IL 60614, U.S.A
| | - Xiao-Wu Qu
- Department of Pathology, Children's Memorial Hospital, 2300 Childrens's Plaza, Chicago, IL 60614, U.S.A
| | - Michael Caplan
- Department of Pediatrics, Evanston Hospital, Northwestern University, Chicago, IL 60614, U.S.A
| | - Frank Gonzalez-Crussi
- Department of Pathology, Children's Memorial Hospital, 2300 Childrens's Plaza, Chicago, IL 60614, U.S.A
| | - Wei Hsueh
- Department of Pathology, Children's Memorial Hospital, 2300 Childrens's Plaza, Chicago, IL 60614, U.S.A
- Author for correspondence:
| |
Collapse
|
43
|
Yeh KY, Yeh M, Glass J, Granger DN. Rapid activation of NF-kappaB and AP-1 and target gene expression in postischemic rat intestine. Gastroenterology 2000; 118:525-34. [PMID: 10702203 DOI: 10.1016/s0016-5085(00)70258-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The molecular mechanisms underlying intestinal mucosal damage-repair processes induced by ischemia-reperfusion (IR) remain unknown. We determined nuclear factor-kappaB (NF-kappaB) and activator protein 1 (AP-1) activities and the expression of potential target genes relevant to damage-repair events. METHODS Rat jejunal segment was subjected to ischemia for 30 minutes followed by reperfusion for defined times. NF-kappaB and AP-1 activities; mucosal p105, p50, and inhibitor kappaB-alpha (IkappaB-alpha) levels; and c-fos, neurotensin, and ferritin H expression were determined by electrophoretic mobility shift assay and Western and Northern analyses, respectively. RESULTS NF-kappaB and AP-1 activities were significantly elevated from 1 to 12 hours after reperfusion. The activated NF-kappaB in the nuclear extract consisted of solely p50 homodimers. Activation of p50 was associated with a decrease of p105, generation of p50, and increased phosphorylation and degradation of IkappaB-alpha. The activated AP-1 contained c-fos but not c-jun, fosB, and Fra-1. Reperfusion induced a transient elevation of c-fos, prolonged increase of neurotensin, and early reduction followed by recovery of ferritin H messenger RNA. CONCLUSIONS The intestine shows organ-specific responses to IR, characterized by prolonged NF-kappaB and AP-1 activation involving NF-kappaB p50 dimers and excluding AP-1 c-jun protein. Degradation of the IkappaB-gamma component of p105 and partial reduction IkappaB-alpha selectively activate p50/p50 dimers. Temporal patterns of target gene expression reflect functional relevance to mucosal damage-repair processes after IR.
Collapse
Affiliation(s)
- K Y Yeh
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | |
Collapse
|
44
|
De Plaen IG, Tan XD, Chang H, Wang L, Remick DG, Hsueh W. Lipopolysaccharide activates nuclear factor kappaB in rat intestine: role of endogenous platelet-activating factor and tumour necrosis factor. Br J Pharmacol 2000; 129:307-14. [PMID: 10694237 PMCID: PMC1571839 DOI: 10.1038/sj.bjp.0703055] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. We examined the effect of lipopolysaccharide (LPS), a cell wall constituent of Gram negative bacteria, on nuclear factor kappaB (NF-kappaB) activation in the intestine and the roles of endogenous platelet-activating factor (PAF), tumour necrosis factor-alpha (TNF) and neutrophils. We also compared the time course of NF-kappaB activation in response to PAF and LPS. 2. Ileal nuclear extracts from LPS (8 mg kg(-1), IV)-injected rats were assayed for NF-kappaB-DNA-binding activity and identification of the subunits. Some rats were pretreated with WEB2170 (a PAF receptor antagonist), anti-TNF antibody, or anti-neutrophil antiserum. NF-kappaB p65 was localized by immunohistochemistry. An additional group was challenged with PAF (2 microg kg(-1), IV) for comparison. 3. LPS activates intestinal NF-kappaB, both as p50-p50 and p50-p65 dimers within 15 min, and the effect peaks at 2 h. The effect is slower and more sustained than that of PAF, which peaks at 30 min. Activated NF-kappaB was immunolocalized within epithelial and lamina propria cells. LPS effect was reduced by 41, 37 and 44%, respectively, in animals pretreated with WEB2170, anti-TNF antibody, or anti-neutrophil antiserum (P<0.05). 4. LPS activates intestinal NF-kappaB in vivo and neutrophil activation is involved in its action. The LPS effect is mediated by both endogenous PAF and TNF.
Collapse
Affiliation(s)
- Isabelle G De Plaen
- Department of Pediatrics (Neonatology), Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Xiao-Di Tan
- Department of Pathology, Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Hong Chang
- Department of Pathology, Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Liya Wang
- Department of Pediatrics (Neonatology), Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
| | - Daniel G Remick
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, MI 48109, U.S.A
| | - Wei Hsueh
- Department of Pathology, Children's Memorial Hospital, Northwestern University Medical School, Chicago, Illinois, IL 60614, U.S.A
- Author for correspondence:
| |
Collapse
|
45
|
Sakaguchi T, Nakamura S, Suzuki S, Oda T, Ichiyama A, Baba S, Okamoto T. Participation of platelet-activating factor in the lipopolysaccharide-induced liver injury in partially hepatectomized rats. Hepatology 1999; 30:959-67. [PMID: 10498648 DOI: 10.1002/hep.510300414] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Platelet-activating factor (PAF) has been shown to be an important mediator in the pathogenesis of lipopolysaccharide (LPS)-induced liver injury in regenerating rat livers. Both LPS and PAF activate nuclear factor-kappa B (NF-kappaB), a key transcription factor for tumor necrosis factor-alpha (TNF-alpha) and cytokine-induced neutrophil chemoattractant (CINC). The aim of this study is to investigate how PAF participates in the LPS-induced and NF-kappaB-mediated regulation of TNF-alpha and CINC in regenerating rat livers. LPS (1.5 mg/kg) was intravenously administered into 70% hepatectomized rats and sham-operated rats 48 hours postoperatively. LPS administration caused a high mortality rate, scattered necrosis in the liver with infiltration of CINC-positive neutrophils, and a continuous CINC messenger RNA up-regulation and activation of NF-kappaB in the liver only in hepatectomized rats. These phenomena were all effectively prevented by pretreatment and posttreatment with a PAF receptor antagonist, TCV-309. Hepatectomized rats showed NF-kappaB staining in hepatocytes, Kupffer cells, and neutrophils around necrosis 4 hours after the LPS injection, representing the activation of this factor in these cells. Based on these results, we propose that PAF contributes to continuous CINC up-regulation and NF-kappaB activation via accumulation and activation of neutrophils, and thereby is involved in LPS-induced liver injury in regenerating rat livers.
Collapse
Affiliation(s)
- T Sakaguchi
- Department of Surgery II, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Merendino N, Dwinell MB, Varki N, Eckmann L, Kagnoff MF. Human intestinal epithelial cells express receptors for platelet-activating factor. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:G810-8. [PMID: 10516147 DOI: 10.1152/ajpgi.1999.277.4.g810] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
The intestinal epithelium produces and responds to cytokines and lipid mediators that play a key role in the induction and regulation of mucosal inflammation. The lipid mediator platelet-activating factor (PAF) can be produced and degraded by the human intestinal epithelium and is known to mediate a range of proinflammatory and other biological effects in the intestinal mucosa. In the studies herein, we assessed whether or not human intestinal epithelial cells express cell surface or intracellular PAF receptors (PAF-R), whether expression of these receptors can be regulated, and whether human intestinal epithelial cells respond to PAF. Several human colon epithelial cell lines (HT-29, Caco-2, T84, HCT-8, HCA-7, I407, and LS-174T) were shown by RT-PCR to constitutively express mRNA for PAF-R. In addition, PAF-R expression was demonstrated by immunoblot analysis and PAF-R was shown to be constitutively expressed on the cell surface of several of these cell lines, as assessed by flow cytometry. PAF-R expression by human colon epithelial cells was upregulated by stimulation with retinoic acid but not by stimulation with PAF, proinflammatory agonists (tumor necrosis factor-alpha, interleukin-1, interferon-gamma), or transforming growth factor-alpha. PAF-R on intestinal epithelial cells were functional, as PAF stimulation of the cells increased tyrosine phosphorylation of several cellular proteins, including proteins of 75 and 125 kDa, and this response was blocked by a PAF-R antagonist. Consistent with the findings using cell lines, PAF-R were also constitutively expressed by normal human colon and small intestinal epithelium in vivo, as shown by immunohistology. The constitutive and regulated expression of functional PAF-R by human intestinal epithelium suggests PAF produced by the intestinal epithelial cells or cells underlying the epithelium has autocrine or paracrine effects on intestinal epithelial cells.
Collapse
Affiliation(s)
- N Merendino
- Department of Medicine, University of California San Diego, La Jolla, California 92093-0623, USA
| | | | | | | | | |
Collapse
|
47
|
Sakaguchi T, Nakamura S, Suzuki S, Oda T, Ichiyama A, Baba S. Acute portal hypertension increases ileal vulnerability to platelet-activating factor in rats. J Surg Res 1999; 86:116-22. [PMID: 10452877 DOI: 10.1006/jsre.1999.5697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Patients with portal hypertension can easily develop sepsis of enteric origin after suffering severe trauma and hemorrhagic shock. Platelet-activating factor (PAF) is one of the key mediators of such stress. The aim of this study was to investigate whether portal hypertension increases the vulnerability of the ileum to PAF. MATERIALS AND METHODS Seven days after surgery, PAF (1.5 microg/kg) was intravenously injected into portal stenosis (PS) rats and sham-operated rats. The levels of tumor necrosis factor-alpha (TNF-alpha), cytokine-induced neutrophil chemoattractant (CINC), and endotoxin in portal plasma were determined. The levels of PAF receptor (PAFR), TNF-alpha, and CINC mRNA in the ileum were also investigated. RESULTS After PAF administration, PS rats showed (1) significantly higher portal plasma levels of TNF-alpha, CINC, and endotoxin; (2) higher histological damage scores in the ileum; (3) more infiltrating neutrophils in the ileum; and (4) a significantly higher mortality rate than sham-operated rats (P < 0.01). However, PAFR mRNA levels were similar in the two groups. The CINC mRNA level in the ileum of PS rats was increased from 1 to 4 h after PAF administration, while that of the sham-operated rats was transiently increased at 1 h. CONCLUSIONS Portal hypertension increases the vulnerability of the ileum to PAF. These findings suggest that conditions which causes PAF production may be dangerous in patients with portal hypertension.
Collapse
Affiliation(s)
- T Sakaguchi
- Department of Surgery II, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | | | | | | | |
Collapse
|