1
|
Sui C, Wang B, Zhao Y, Guo Y, Zhu J, Yu F, Zhou X, Bu X, Zhang J. Establishment of an inflammatory cytokine-based predictive model for the onset of prolonged postoperative ileus after radical gastrectomy: a prospective cohort study. Front Immunol 2025; 16:1552944. [PMID: 40196130 PMCID: PMC11973057 DOI: 10.3389/fimmu.2025.1552944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Background Prolonged postoperative ileus (PPOI) is a common postoperative abdominal complication and is strongly associated with the inflammatory response. However, there is a lack of effective means to predict PPOI in patients with gastric cancer. Methods 222 patients underwent radical gastrectomy at our center were enrolled and divided into the training group and validation cohort. Receiver operating characteristic (ROC) curve analysis and univariate and multivariable logistic regression models were performed to help filter variables for inclusion in the predictive model. And then a nomogram for PPOI was established. The area under the ROC curve (AUC) was calculated to assess the prediction accuracy. Diagnostic calibration curves were used to assess the goodness-of-fit of the nomogram. Decision Curve Analysis (DCA) was applied to evaluate its clinical utility. Results Significant increase of IL-6, IL-10, TNF-α, and CRP on the first postoperative day were found in PPOI patients after surgery. Univariate and multivariate analysis demonstrated that age ≥ 65, IL-6, and IL-10 were independent predictive factors for PPOI. We subsequently developed a prediction nomogram of PPOI which included age, IL-6, IL-10, and TNF-α. Further verification by the training and validation groups demonstrated the good predictive efficacy of our model, as well as favorable clinical benefits. Conclusions We developed a novel and easy-to-use prediction nomogram for gastric cancer, which was primarily based on the postoperative level of inflammatory mediators. This model provided further clarification of the exact relationship between inflammatory factors and the occurrence of PPOI, and help us clinically identify the high-risk groups of PPOI for the purpose of early intervention.
Collapse
Affiliation(s)
- Chao Sui
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - BeiBei Wang
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - Yu Zhao
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - YunTian Guo
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - JinXin Zhu
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - Feng Yu
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - XiaoDong Zhou
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - XueFeng Bu
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - Jie Zhang
- Department of General Surgery, Jiangsu University Affiliated People’s Hospital, Zhenjiang, China
- Department of General Surgery, Zhenjiang First People’s Hospital, Zhenjiang, China
| |
Collapse
|
2
|
Breßer M, Siemens KD, Schneider L, Lunnebach JE, Leven P, Glowka TR, Oberländer K, De Domenico E, Schultze JL, Schmidt J, Kalff JC, Schneider A, Wehner S, Schneider R. Macrophage-induced enteric neurodegeneration leads to motility impairment during gut inflammation. EMBO Mol Med 2025; 17:301-335. [PMID: 39762650 PMCID: PMC11822118 DOI: 10.1038/s44321-024-00189-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Current studies pictured the enteric nervous system and macrophages as modulators of neuroimmune processes in the inflamed gut. Expanding this view, we investigated the impact of enteric neuron-macrophage interactions on postoperative trauma and subsequent motility disturbances, i.e., postoperative ileus. In the early postsurgical phase, we detected strong neuronal activation, followed by transcriptional and translational signatures indicating neuronal death and synaptic damage. Simultaneously, our study revealed neurodegenerative profiles in macrophage-specific transcriptomes after postoperative trauma. Validating the role of resident and monocyte-derived macrophages, we depleted macrophages by CSF-1R-antibodies and used CCR2-/- mice, known for reduced monocyte infiltration, in POI studies. Only CSF-1R-antibody-treated animals showed decreased neuronal death and lessened synaptic decay, emphasizing the significance of resident macrophages. In human gut samples taken early and late during abdominal surgery, we substantiated the mouse model data and found reactive and apoptotic neurons and dysregulation in synaptic genes, indicating a species' overarching mechanism. Our study demonstrates that surgical trauma activates enteric neurons and induces neurodegeneration, mediated by resident macrophages, introducing neuroprotection as an option for faster recovery after surgery.
Collapse
Affiliation(s)
- Mona Breßer
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kevin D Siemens
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Patrick Leven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristin Oberländer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Elena De Domenico
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim Schmidt
- Department of General, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
3
|
Duan WQ, Cai MC, Ma QQ, Huang P, Zhang JH, Wei TF, Shang D, Leng AJ, Qu JL. Exploring the chemical components of Kuanchang-Shu granule and its protective effects of postoperative ileus in rats by regulating AKT/HSP90AA1/eNOS pathway. Chin Med 2024; 19:29. [PMID: 38383512 PMCID: PMC10880223 DOI: 10.1186/s13020-024-00892-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/21/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Postoperative ileus (POI) is a common obstruction of intestinal content passage caused by almost all abdominal operations that seriously strokes the quality of life of patients. Kuanchang-Shu granule (KCSG), a classic modified prescription based on "Da-Cheng-Qi Decoction", has obtained satisfactory efficacy in the clinical therapeutics of POI. However, its material basis and holistic molecular mechanism against POI have not been revealed. METHODS The chemical ingredients of KCSG were first characterized by ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-QTOF-MS). Subsequently, an integration strategy of the network pharmacology and molecular docking based on above identified ingredients was performed to unveil the potential targets involved in the treatment of KCSG on POI. Finally, intestinal manipulation induced rat POI model was constructed to verify the efficacy and predicted mechanism of KCSG against POI. RESULTS In total, 246 ingredients mainly including organic acids, flavonoids, quinones, alkaloids, terpenoids, phenylpropanoids and phenols were identified. 41 essential ingredients, 24 crucial targets as well as 15 relevant signaling pathways were acquired based on network pharmacology analysis. Pharmacodynamic research showed that KCSG treatment could protect intestinal histological damage, promote the recovery of measurement of gastrointestinal transit disorder and inhibit the secretion of myeloperoxidase in the distal ileum tissues. The up-regulated expression of p-AKT and down-regulated expression of p-eNOS and HSP9OAA1 predicted by molecular docking and validated by western blotting showed that AKT/eNOS/HSP90AA1 pathway may be one of the crucial mechanisms that mediates the protective effect of KCSG.
Collapse
Affiliation(s)
- Wen-Qian Duan
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Ming-Chen Cai
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
| | - Qi-Qi Ma
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
| | - Peng Huang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Jia-Hui Zhang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Tian-Fu Wei
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Ai-Jing Leng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China.
| | - Jia-Lin Qu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, China.
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9, South Road of Lvshun, Dalian, 116044, China.
| |
Collapse
|
4
|
Yang R, Wang X, Liu H, Chen J, Tan C, Chen H, Wang X. Egr-1 is a key regulator of the blood-brain barrier damage induced by meningitic Escherichia coli. Cell Commun Signal 2024; 22:44. [PMID: 38233877 PMCID: PMC10795328 DOI: 10.1186/s12964-024-01488-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Bacterial meningitis remains a leading cause of infection-related mortality worldwide. Although Escherichia coli (E. coli) is the most common etiology of neonatal meningitis, the underlying mechanisms governing bacterial blood-brain barrier (BBB) disruption during infection remain elusive. We observed that infection of human brain microvascular endothelial cells with meningitic E. coli triggers the activation of early growth response 1 (Egr-1), a host transcriptional activator. Through integrated chromatin immunoprecipitation sequencing and transcriptome analysis, we identified Egr-1 as a crucial regulator for maintaining BBB integrity. Mechanistically, Egr-1 induced cytoskeletal changes and downregulated tight junction protein expression by directly targeting VEGFA, PDGFB, and ANGPTL4, resulting in increased BBB permeability. Meanwhile, Egr-1 also served as a master regulator in the initiation of neuroinflammatory response during meningitic E. coli infection. Our findings support an Egr-1-dependent mechanism of BBB disruption by meningitic E. coli, highlighting a promising therapeutic target for bacterial meningitis.
Collapse
Affiliation(s)
- Ruicheng Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Xinyi Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Hulin Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jiaqi Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China.
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China.
| |
Collapse
|
5
|
Shi X, Wang Y, Liu H, Han R. Targeting Hub Genes Involved in Muscle Injury Induced by Jumping Load Based on Transcriptomics. DNA Cell Biol 2023; 42:498-506. [PMID: 37339448 DOI: 10.1089/dna.2022.0285] [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] [Indexed: 06/22/2023] Open
Abstract
The purpose of this study was to find hub genes that may play key roles in skeletal muscle injury induced by jumping load. Twelve female Sprague Dawley rats were divided into the normal control (NC) group and the jumping-induced muscle injury (JI) group. After 6 weeks of jumping, transmission electron microscopy, hematoxylin-eosin staining, transcriptomics sequencing and genes analysis, interaction network prediction of multiple proteins, real-time PCR detection, and Western blotting were performed on gastrocnemius muscles from NC and JI groups. As compared with NC rats, excessive jumping can result in notable structural damage and inflammatory infiltration in JI rats. A total of 112 differentially expressed genes were confirmed in NC rats versus JI rats, with 59 genes upregulated and 53 genes downregulated. Using the online String database, four hub genes in the transcriptional regulatory network were targeted, including FOS, EGR1, ATF3, and NR4A3. All expression levels of FOS, EGR1, ATF3, and NR4A3 mRNAs were decreased in JI rats compared with NC rats (p < 0.05 or p < 0.01). All expression levels of c-Fos, EGR1, ATF3, and NOR1 proteins were upregulated in JI rats (p < 0.01, p < 0.05, p > 0.05, and p < 0.01, respectively). Collectively, these findings indicate that FOS, EGR1, ATF3, and NR4A3 genes may be functionally important in jumping-induced muscle injury.
Collapse
Affiliation(s)
- Xiaolan Shi
- Wushu College, Henan University, Kaifeng, China
| | - Yijie Wang
- School of Physical Education and Sport, Henan University, Kaifeng, China
| | - Haitao Liu
- School of Physical Education and Sport, Henan University, Kaifeng, China
- Sports Reform and Development Research Center, Henan University, Kaifeng, China
| | - Rui Han
- School of Physical Education and Sport, Henan University, Kaifeng, China
- Sports Reform and Development Research Center, Henan University, Kaifeng, China
| |
Collapse
|
6
|
Yuan PQ, Wu SV, Wang L, Taché Y. The ghrelin agonist, HM01 activates central vagal and enteric cholinergic neurons and reverses gastric inflammatory and ileus responses in rats. Neurogastroenterol Motil 2023; 35:e14561. [PMID: 36942655 DOI: 10.1111/nmo.14561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Electrical vagal stimulation alleviates abdominal surgery (AS)-induced intestinal inflammation. Ghrelin receptors (GHS-Rs) are expressed in the brain and peripheral tissues. We investigated the influence of HM01, an orally active ghrelin agonist crossing the blood-brain barrier, on AS-induced gastric inflammation and emptying (GE) in rats. METHODS HM01 (6 mg/kg) or saline pretreatment was administered per orally (po) or intraperitoneally (ip). We assessed GE, gastric cytokine mRNA, and Fos positive cells in the dorsal motor nucleus of the vagus (DMN) and gastric corpus myenteric plexus (MP) in sham (anesthesia alone) and AS groups. The transcripts of GHS-R1 variants were determined in the medulla oblongata and gastric corpus of naïve rats. KEY RESULTS In vehicle pretreated rats, HM01 (ip) significantly increased the number of Fos immunoreactive cells in the MP and DMN in 55% and 52% of cholinergic neurons respectively. Hexamethonium did not modify HM01-induced Fos expression in the DMN while reducing it in the MP by 2-fold with values still significantly higher than that in control groups. AS upregulated gastric IL-1β and TNFα expression and inhibited GE by 66.6%. HM01 (po) abolished AS-induced gastric ileus and increased cytokine expression and elevated IL-10 by 4.0-fold versus vehicle/sham. GHS-R1a mRNA level was 5.4-fold higher than the truncated GHS-R1b isoform in the brain medulla and 40-fold higher in the gastric submucosa/muscle layers than in the mucosa. CONCLUSIONS AND INFERENCE Peripheral HM0 activates central vagal and myenteric cholinergic pathways that may influence both central and peripheral targets to prevent AS-induced gastric inflammatory and ileus.
Collapse
Affiliation(s)
- Pu-Qing Yuan
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| | - S Vincent Wu
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Lixin Wang
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| | - Yvette Taché
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
7
|
Arnauts K, Sudhakar P, Verstockt S, Lapierre C, Potche S, Caenepeel C, Verstockt B, Raes J, Vermeire S, Sabino J, Verfaillie C, Ferrante M. Microbiota, not host origin drives ex vivo intestinal epithelial responses. Gut Microbes 2022; 14:2089003. [PMID: 35758256 PMCID: PMC9235885 DOI: 10.1080/19490976.2022.2089003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Microbial dysbiosis is an established finding in patients with inflammatory bowel disease (IBD), but host-microbial interactions are poorly understood. We aimed to unravel the effect of microbiota exposure on intestinal epithelial cells. Confluent Transwell® organoid monolayers of eight UC patients and eight non-IBD controls were co-cultured for six hours with microbiota (3x108 cells) of UC patients or a healthy volunteer (HV), in the presence or absence of an inflammatory cytokine mix. Transepithelial electrical resistance (TEER), fluorescein isothiocyanate (FITC) dextran measurements, and RNA sequencing were performed on epithelial cells, and 16S rRNA sequencing on microbiota samples before and after co-culture. Transcriptomic response following microbiota exposure was not different between epithelial cells from UC patients or non-IBD controls. Following UC microbiota exposure, but not HV microbiota, a strong decrease in epithelial barrier integrity was observed in both UC and HV epithelial cells by TEER and FITC dextran measurements. Exposure of inflamed epithelium to UC microbiota induced transcriptomic stress pathways including activation of EGR1, MAPK and JAK/STAT signaling, as well as AP-1 family and FOSL transcripts. Stress responses after HV microbiota stimulation were milder. We conclude that not the epithelial cell origin (UC versus non-IBD) but the microbial donor drives transcriptomic responses, as exposure to UC microbiota was sufficient to induce stress responses in all epithelial cells. Further research on therapies to restore the microbial balance, to remove the constant trigger of dysbiosis, is required.
Collapse
Affiliation(s)
- Kaline Arnauts
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Development and Regeneration, Stem Cell Institute Leuven (SCIL), KU Leuven, Leuven, Belgium
| | - Padhmanand Sudhakar
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Sare Verstockt
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Cynthia Lapierre
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Selina Potche
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Clara Caenepeel
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - João Sabino
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute Leuven (SCIL), KU Leuven, Leuven, Belgium
| | - Marc Ferrante
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium,CONTACT Marc Ferrante Department of Gastroenterology and Hepatology University Hospitals Leuven, Herestraat 49, Leuven3000, Belgium
| |
Collapse
|
8
|
Liu L, Liu Y, Guo X, Jin X, Yan W, Lin B, Cai T, Wei Y. Activation of p38 mitogen-activated protein kinase pathway by lipopolysaccharide aggravates postoperative ileus in colorectal cancer patients. J Gastroenterol Hepatol 2022; 37:518-530. [PMID: 34907602 DOI: 10.1111/jgh.15760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/04/2021] [Accepted: 12/04/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM Patients undergoing abdominal surgery can develop postoperative ileus (POI). Inflammation of the intestinal muscularis following intestinal manipulation may be caused by displaced bacteria or lipopolysaccharide (LPS). The aim of this study was to investigate the relationship between gut microbiota, LPS, and POI in colorectal cancer (CRC) patients and explore underlying mechanisms of LPS-triggered POI. METHODS Sixty CRC patients undergoing colorectal resection were included. Bacterial communities from fecal samples were characterized by 16S rRNA gene sequencing, and fecal LPS levels were determined by Limulus amebocyte lysate assay. Mice were used to mechanistically investigate the causal relationship between microbiota, LPS, and POI. RESULTS We discovered that CRC patients who developed prolonged POI (PPOI) had a unique pro-inflammatory gut microbial composition during the perioperative period. The highest proportions of Gram-negative bacteria at the genus level were Escherichia-Shigella and Bacteroides; the abundance of Escherichia-Shigella was higher throughout the perioperative period. Fecal LPS levels were significantly higher in patients with PPOI. In mice treated with an antibiotic cocktail, intestinal muscularis inflammation and intestinal dysfunction were significantly improved. Inflammation and dysfunction were significantly reduced in mice treated with polymyxin B, but were worsened by treatment with LPS. Moreover, LPS upregulated p38 phosphorylation in mice, and treatment with an inhibitor of p38 (SB203580) significantly alleviated intestinal inflammation and dysmotility. CONCLUSION Lipopolysaccharide increases intestinal muscularis inflammation via activation of p38 signaling, which aggravates POI. Removing bacterial sources of LPS during the perioperative period is promising for the prophylactic treatment of PPOI.
Collapse
Affiliation(s)
- Lujia Liu
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yang Liu
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China
| | - Xiao Guo
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangren Jin
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Yan
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baiqiang Lin
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ting Cai
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Yunwei Wei
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China
| |
Collapse
|
9
|
Wang RS, Lembo AJ, Kaptchuk TJ, Cheng V, Nee J, Iturrino J, Rao M, Loscalzo J, Silvester JA, Hall KT. Genomic Effects Associated With Response to Placebo Treatment in a Randomized Trial of Irritable Bowel Syndrome. FRONTIERS IN PAIN RESEARCH 2022; 2:775386. [PMID: 35295415 PMCID: PMC8915627 DOI: 10.3389/fpain.2021.775386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Irritable bowel syndrome (IBS), a functional pain disorder of gut-brain interactions, is characterized by a high placebo response in randomized clinical trials (RCTs). Catechol-O-methyltransferase (COMT) rs4680, which encodes high-activity (val) or low-activity (met) enzyme variants, was previously associated with placebo response to sham-acupuncture in an IBS RCT. Examining COMT effects and identifying novel genomic factors that influence response to placebo pills is critical to identifying underlying mechanisms and predicting and managing placebos in RCTs. Methods: Participants with IBS (N = 188) were randomized to three placebo-related interventions, namely, double-blind placebo (DBP), open-label placebo (OLP), or simply trial enrollment without placebo treatment [no placebo (i.e., no pill) treatment control (NPC)], for 6 weeks. COMT rs4680, gene-set, and genome-wide suggestive (p < 10-5) loci effects on irritable bowel symptom severity score (IBS-SSS) across all participants were examined. Results: Participants with IBS homozygous for rs4680 met (met/met) had the greatest improvement across all arms, with significantly greater improvement compared to val/val in DBP (beta (SE), -89.4 (42.3); p = 0.04). Twelve genome-wide suggestive loci formed a gene regulatory network highly connected to EGR1, a transcription factor involved in placebo-related processes of learning, memory, and response to stress and reward. EGR1 gene expression in peripheral blood mononuclear cells (PBMC) was significantly reduced at the endpoint across all treatment arms (log fold-change, -0.15; p = 0.02). Gene-set enrichment analysis returned three genome-wide significant ontology terms (GO:0032968, GO:0070934, and GO:0070937) linked to transcription regulation and GO:0003918 associated with DNA topoisomerase regulation. Conclusion: These results suggest common molecular mechanisms in response to varying forms of placebo that may inform personalized IBS treatment and placebo response prediction. Clinical Trial Registration: ClinicalTrials.gov, Identifier: NCT0280224.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Department of Medicine, Brigham Women's Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Anthony J. Lembo
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Ted J. Kaptchuk
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of General Medicine Primary Care, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Vivian Cheng
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Judy Nee
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Johanna Iturrino
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Joseph Loscalzo
- Department of Medicine, Brigham Women's Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Jocelyn A. Silvester
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Celiac Disease Program, Boston Children's Hospital, Boston, MA, United States
| | - Kathryn T. Hall
- Department of Medicine, Brigham Women's Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
10
|
Endo M, Oikawa T, Tonooka M, Hanawa T, Odaguchi H, Hori M. Hangekobokuto, a traditional Japanese herbal medicine, ameliorates postoperative ileus through its anti-inflammatory action. J Smooth Muscle Res 2022; 58:78-88. [PMID: 36216552 PMCID: PMC9537061 DOI: 10.1540/jsmr.58.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background/Aims: Gastroprokinetic agents are used for patients with
postoperative ileus (POI), and the Japanese traditional herbal medicine daikenchuto (DKT)
is one such agent used in the clinical setting. POI is caused by inflammation. DKT and
rikkunshito have anti-inflammatory abilities in addition to their gastroprokinetic
effects. The efficacy of Kampo formulations, including hangekobokuto (HKT), in patients
with POI has been reported recently. Several authors have described the efficacy of
honokiol, the primary component of Magnoliae Cortex, in HKT in mouse
models of POI. We therefore analyzed the effect of HKT on POI model mice to determine the
similarities in the mechanism of action between HKT and DKT. Methods: HKT was administered orally to each mouse before and after
intestinal manipulation was performed on the distal ileum. The gastrointestinal transit
in vivo, leukocyte infiltration, and levels of inflammatory mediators,
such as cytokines and chemokines, were analyzed. Results: HKT significantly inhibited the infiltration of neutrophils and
macrophages and led to the recovery of delayed intestinal transit. In addition, it
significantly decreased inducible nitric oxide synthase (iNOS) as well as honokiol levels,
suggesting anti-inflammatory activity. However, it did not inhibit the increase in levels
of interleukin (IL)-1beta and IL-6, which are related to iNOS induction. In contrast, HKT
increased levels of nerve growth factor (NGF) and suppressed those of nuclear factor-κB
(NFκB), which are related to iNOS induction, suggesting the possibility of a neuronal
anti-inflammatory mechanism. Conclusions: HKT exerted a POI-relieving effect similar to DKT in a murine
POI model, and findings suggest that it may exert its anti-inflammatory activity through
NGF.
Collapse
Affiliation(s)
- Mari Endo
- Department of Clinical Research, Oriental Medicine Research
Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8642, Japan
| | - Tetsuro Oikawa
- Center for Kampo Medicine, Tokyo Medical University Hospital,
6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Miki Tonooka
- Graduate School of Infection Control Sciences, Kitasato
University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8642, Japan
| | - Toshihiko Hanawa
- Department of Clinical Research, Oriental Medicine Research
Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8642, Japan
| | - Hiroshi Odaguchi
- Department of Clinical Research, Oriental Medicine Research
Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8642, Japan,Oriental Medicine, Doctoral Program of Medical Science,
Kitasato University Graduate School, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8642,
Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of
Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo
113-8657, Japan
| |
Collapse
|
11
|
Zou K, Chen W, Dai J, Mo P, Yu C, Xu J, Wu S, Zhuo R, Su G. Steroid Receptor Coactivator-3 Is Required for Inhibition of the Intestinal Muscularis Inflammatory Response of Postoperative Ileus in Mice. Inflammation 2021; 44:1145-1159. [PMID: 33398542 DOI: 10.1007/s10753-020-01409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
Inflammation theory has suggested that the pathogenesis of postoperative ileus (POI) involves the steroid receptor coactivator-3 (SRC-3). Therefore, we investigated the role of SRC-3 in the muscles of the small intestine using a mouse POI model. Here, we reported that intestinal manipulation (IM) significantly reduced the extent of phenol red migration in the entire gastrointestinal tract, and the calculated geometric center (GC) value in wild-type (WT) mice at 24 h after surgery was higher than that in the knockout (KO) mice and in the sham-operated control group. The expression of SRC-3 was upregulated in the mouse intestinal muscularis at 24 h after surgical manipulation, and the mRNA and protein levels of inflammatory cytokines were upregulated compared with those in the control group. At 24 h after IM, the number of neutrophils in the experimental group was significantly higher than that in the control group; in the IM group, the number of neutrophils in the SRC-3-/- mice was markedly higher than that in the WT mice. At 24 h after IM, the myeloperoxidase (MPO) activity in the experimental group was significantly higher than that in the control group. In the IM group, the MPO activity of the SRC-3-/- mice was markedly higher than that of the WT mice. In summary, proinflammatory cytokines, the number of neutrophils, and the MPO activity were significantly increased in the muscularis of the jejunum and ileum of KO mice after IM compared with those of the WT mice, indicating that SRC-3 might play a protective role in POI.
Collapse
MESH Headings
- Animals
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Gastrointestinal Motility
- Ileus/etiology
- Ileus/immunology
- Ileus/metabolism
- Ileus/physiopathology
- Inflammation Mediators/metabolism
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intestine, Small/physiopathology
- Jejunum/immunology
- Jejunum/metabolism
- Jejunum/physiopathology
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth/immunology
- Muscle, Smooth/metabolism
- Muscle, Smooth/physiopathology
- Neutrophil Infiltration
- Nuclear Receptor Coactivator 3/genetics
- Nuclear Receptor Coactivator 3/metabolism
- Peroxidase/metabolism
- Postoperative Complications/etiology
- Postoperative Complications/immunology
- Postoperative Complications/metabolism
- Postoperative Complications/physiopathology
- Tissue Culture Techniques
- Mice
Collapse
Affiliation(s)
- Kang Zou
- Department of Gastrointestinal Surgery, Ward 3 Areas of Cancer Center, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, China
- Department of Gastrointestinal Surgery, Ward 3 Areas of Cancer Center, Cancer Hospital, Teaching Hospital of Fujian Medical University, Xiamen, 361003, Fujian, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Gannan Medical College, Ganzhou, 341000, China
| | - Wenbo Chen
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
- Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Jun Dai
- Department of Gastrointestinal Surgery, Ward 3 Areas of Cancer Center, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, China
| | - Pingli Mo
- Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Chundong Yu
- Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sangang Wu
- Department of Radiation Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, 361003, China
| | - Rengong Zhuo
- School of Medicine, Xiamen University, Xiang'an District, Xiamen, 361102, China.
| | - Guoqiang Su
- Department of Gastrointestinal Surgery, Ward 3 Areas of Cancer Center, Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, China.
- Department of Gastrointestinal Surgery, Ward 3 Areas of Cancer Center, Cancer Hospital, Teaching Hospital of Fujian Medical University, Xiamen, 361003, Fujian, China.
| |
Collapse
|
12
|
Zhu X, Li F, Wang M, Su H, Wu X, Qiu H, Zhou W, Shan C, Wang C, Wei L. Integrated Analysis of Omics Data Reveal AP-1 as a Potential Regulation Hub in the Inflammation-Induced Hyperalgesia Rat Model. Front Immunol 2021; 12:672498. [PMID: 34122430 PMCID: PMC8194263 DOI: 10.3389/fimmu.2021.672498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022] Open
Abstract
Inflammation-associated chronic pain is a global clinical problem, affecting millions of people worldwide. However, the underlying mechanisms that mediate inflammation-associated chronic pain remain unclear. A rat model of cutaneous inflammation induced by Complete Freund's Adjuvant (CFA) has been widely used as an inflammation-induced pain hypersensitivity model. We present the transcriptomics profile of CFA-induced inflammation in the rat dorsal root ganglion (DRG) via an approach that targets gene expression, DNA methylation, and post-transcriptional regulation. We identified 418 differentially expressed mRNAs, 120 differentially expressed microRNAs (miRNAs), and 2,670 differentially methylated regions (DMRs), which were all highly associated with multiple inflammation-related pathways, including nuclear factor kappa B (NF-κB) and interferon (IFN) signaling pathways. An integrated analysis further demonstrated that the activator protein 1 (AP-1) network, which may act as a regulator of the inflammatory response, is regulated at both the transcriptomic and epigenetic levels. We believe our data will not only provide drug screening targets for the treatment of chronic pain and inflammation but will also shed light on the molecular network associated with inflammation-induced hyperalgesia.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Li
- The Yancheng Clinical College of XuZhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
| | - Miqun Wang
- Department of Anesthesiology, Qingdao Women and Children's Hospital, Qingdao, China
| | - Huibin Su
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Xuedong Wu
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Haiyan Qiu
- Department of Anesthesiology, The First People's Hospital of Yancheng, Yancheng, China
| | - Wang Zhou
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China.,Graduate School of Nantong University, Nantong, China
| | - Chunli Shan
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China.,Graduate School of Nantong University, Nantong, China
| | - Cancan Wang
- Graduate School of Nantong University, Nantong, China
| | - Lei Wei
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| |
Collapse
|
13
|
Enderes J, Mallesh S, Schneider R, Hupa KJ, Lysson M, Schneiker B, Händler K, Schlotmann B, Günther P, Schultze JL, Kalff JC, Wehner S. A Population of Radio-Resistant Macrophages in the Deep Myenteric Plexus Contributes to Postoperative Ileus Via Toll-Like Receptor 3 Signaling. Front Immunol 2021; 11:581111. [PMID: 33519804 PMCID: PMC7838642 DOI: 10.3389/fimmu.2020.581111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/27/2020] [Indexed: 11/13/2022] Open
Abstract
Postoperative ileus (POI) is triggered by an innate immune response in the muscularis externa (ME) and is accompanied by bacterial translocation. Bacteria can trigger an innate immune response via toll-like receptor (TLR) activation, but the latter's contribution to POI has been disproved for several TLRs, including TLR2 and TLR4. Herein we investigated the role of double-stranded RNA detection via TLR3 and TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling pathway in POI. POI was induced by small bowel intestinal manipulation in wt, TRIF-/-, TLR3-/-, type I interferon receptor-/- and interferon-β reporter mice, all on C57BL/6 background, and POI severity was quantified by gene expression analysis, gastrointestinal transit and leukocyte extravasation into the ME. TRIF/TLR3 deficiency reduced postoperative ME inflammation and prevented POI. With bone marrow transplantation, RNA-sequencing, flow cytometry and immunohistochemistry we revealed a distinct TLR3-expressing radio-resistant MHCIIhiCX3CR1- IBA-1+ resident macrophage population within the deep myenteric plexus. TLR3 deficiency in these cells, but not in MHCIIhiCX3CR1+ macrophages, reduced cytokine expression in POI. While this might not be an exclusive macrophage-privileged pathway, the TLR3/TRIF axis contributes to proinflammatory cytokine production in MHCIIhiCX3CR1- IBA-1+ macrophages during POI. Deficiency in TLR3/TRIF protects mice from POI. These data suggest that TLR3 antagonism may prevent POI in humans.
Collapse
Affiliation(s)
- Jana Enderes
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Shilpashree Mallesh
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Reiner Schneider
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Kristof J Hupa
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Mariola Lysson
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Bianca Schneiker
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Kristian Händler
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Balthasar Schlotmann
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Patrick Günther
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, Division of Immune Pathophysiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
14
|
Alam A, Imam N, Siddiqui MF, Ali MK, Ahmed MM, Ishrat R. Human gene expression profiling identifies key therapeutic targets in tuberculosis infection: A systematic network meta-analysis. INFECTION GENETICS AND EVOLUTION 2021; 87:104649. [PMID: 33271338 DOI: 10.1016/j.meegid.2020.104649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/14/2022]
|
15
|
He R, Zhao Z, Yang Y, Liang X. Using bioinformatics and metabolomics to identify altered granulosa cells in patients with diminished ovarian reserve. PeerJ 2020; 8:e9812. [PMID: 32923184 PMCID: PMC7457930 DOI: 10.7717/peerj.9812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 11/20/2022] Open
Abstract
Background During fertility treatment, diminished ovarian reserve (DOR) is a challenge that can seriously affect a patient's reproductive potential. However, the pathogenesis of DOR is still unclear and its treatment options are limited. This study aimed to explore DOR's molecular mechanisms. Methods We used R software to analyze the mRNA microarray dataset E-MTAB-391 downloaded from ArrayExpress, screen for differentially expressed genes (DEGs), and perform functional enrichment analyses. We also constructed the protein-protein interaction (PPI) and miRNA-mRNA networks. Ovarian granulosa cells (GCs) from women with DOR and the control group were collected to perform untargeted metabolomics analyses. Additionally, small molecule drugs were identified using the Connectivity Map database. Results We ultimately identified 138 DEGs. Our gene ontology (GO) analysis indicated that DEGs were mainly enriched in cytokine and steroid biosynthetic processes. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG), the DEGs were mainly enriched in the AGE (advanced glycation end-product)-RAGE (receptor for AGE) signaling pathway in diabetic complications and steroid biosynthesis. In the PPI network, we determined that JUN, EGR1, HMGCR, ATF3, and SQLE were hub genes that may be involved in steroid biosynthesis and inflammation. miRNAs also played a role in DOR development by regulating target genes. We validated the differences in steroid metabolism across GCs using liquid chromatography-tandem mass spectrometry (LC-MS/MS). We selected 31 small molecules with potentially positive or negative influences on DOR development. Conclusion We found that steroidogenesis and inflammation played critical roles in DOR development, and our results provide promising insights for predicting and treating DOR.
Collapse
Affiliation(s)
- Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Zhongying Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| |
Collapse
|
16
|
Meijerink M, van den Broek TJ, Dulos R, Garthoff J, Knippels L, Knipping K, Harthoorn L, Houben G, Verschuren L, van Bilsen J. Network-Based Selection of Candidate Markers and Assays to Assess the Impact of Oral Immune Interventions on Gut Functions. Front Immunol 2019; 10:2672. [PMID: 31798593 PMCID: PMC6863931 DOI: 10.3389/fimmu.2019.02672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/29/2019] [Indexed: 12/05/2022] Open
Abstract
To assess the safety and efficacy of oral immune interventions, it is important and required by regulation to assess the impact of those interventions not only on the immune system, but also on other organs such as the gut as the porte d'entrée. Despite clear indications that the immune system interacts with several physiological functions of the gut, it is still unknown which pathways and molecules are crucial to assessing the impact of nutritional immune interventions on gut functioning. Here we used a network-based systems biology approach to clarify the molecular relationships between immune system and gut functioning and to identify crucial biomarkers to assess effects on gut functions upon nutritional immune interventions. First, the different gut functionalities were categorized based on literature and EFSA guidance documents. Moreover, an overview of the current assays and methods to measure gut function was generated. Secondly, gut-function related biological processes and adverse events were selected and subsequently linked to the physiological functions of the GI tract. Thirdly, database terms and annotations from the Gene ontology database and the Comparative Toxicogenomics Database (CTD) related to the previously selected gut-function related processes were selected. Next, database terms and annotations were used to identify the pathways and genes involved in those gut functionalities. In parallel, information from CTD was used to identify immune disease related genes. The resulting lists of both gut and immune function genes showed an overlap of 753 genes out of 1,296 gut-function related genes indicating the close gut-immune relationship. Using bioinformatics enrichment tools DAVID and Panther, the identified gut-immune markers were predicted to be involved in motility, barrier function, the digestion and absorption of vitamins and fat, regulation of the digestive system and gastric acid, and protection from injurious or allergenic material. Concluding, here we provide a promising systems biology approach to identify genes that help to clarify the relationships between immune system and gut functioning, with the aim to identify candidate biomarkers to monitor nutritional immune intervention assays for safety and efficacy in the general population. This knowledge helps to optimize future study designs to predict effects of nutritional immune intervention on gut functionalities.
Collapse
Affiliation(s)
| | | | | | | | - Léon Knippels
- Danone Nutricia Research, Utrecht, Netherlands.,Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Karen Knipping
- Danone Nutricia Research, Utrecht, Netherlands.,Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | | | | | | | | |
Collapse
|
17
|
A Functional Polymorphism-Mediated Disruption of EGR1/ADAM10 Pathway Confers the Risk of Sepsis Progression. mBio 2019; 10:mBio.01663-19. [PMID: 31387910 PMCID: PMC6686044 DOI: 10.1128/mbio.01663-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence has indicated that single nucleotide polymorphisms (SNPs) are related to the susceptibility of sepsis and might provide potential evidence for the mechanisms of sepsis. Our recent preliminary study showed that the ADAM10 genetic polymorphism was clinically associated with the development of sepsis, and little is known about the underlying mechanism. The aim of this study was to confirm the association between the ADAM10 promoter rs653765 G→A polymorphism and the progression of sepsis and to discover the underlying mechanism. Clinical data showed that the rs653765 G→A polymorphism was positively correlated with the development of sepsis, as evidenced by a multiple-center case-control association study with a large sample size, and showed that EGR1 and ADAM10 levels were associated well with the different subtypes of sepsis patients. In vitro results demonstrated that the rs653765 G→A variants could functionally modulate ADAM10 promoter activity by altering the binding of the EGR1 transcription factor (TF) to the ADAM10 promoter, affecting the transcription and translation of the ADAM10 gene. Electrophoretic mobility shift assay (EMSA) followed by chromatin immunoprecipitation (ChIP) assay indicated the direct interaction. Functional studies further identified that the EGR1/ADAM10 pathway is important for the inflammatory response. EGR1 intervention in vivo decreased host proinflammatory cytokine secretion and rescued the survival and tissue injury of the mouse endotoxemia model.IMPORTANCE Sepsis is characterized as life-threatening organ dysfunction, with unacceptably high mortality. Evidence has indicated that functional SNPs within inflammatory genes are associated with susceptibility, progression, and prognosis of sepsis. These mechanisms on which these susceptible sites depended often suggest the key pathogenesis and potential targets in sepsis. In the present study, we confirmed that a functional variant acts as an important genetic factor that confers the progression of sepsis in a large sample size and in multiple centers and revealed that the variants modulate the EGR1/ADAM10 pathway and influence the severity of sepsis. We believe that we provide an important insight into this new pathway involving the regulation of inflammatory process of sepsis based on the clinical genetic evidence, which will enhance the understanding of nosogenesis of sepsis and provide the potential target for inflammation-related diseases.
Collapse
|
18
|
Endo M, Hori M, Ozaki H, Oikawa T, Odaguchi H, Hanawa T. Possible anti-inflammatory role of Zingiberis processum rhizoma, one component of the Kampo formula daikenchuto, against neutrophil infiltration through muscarinic acetylcholine receptor activation. J Pharmacol Sci 2018; 137:379-386. [PMID: 30145033 DOI: 10.1016/j.jphs.2018.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Zingiberis processum rhizoma (ZPR) is a major active component of daikenchuto (DKT), which induces anti-inflammatory action by inhibiting macrophage infiltration. However, it is unclear whether ZPR is related to DKT-induced anti-inflammatory action via a reduction of neutrophil infiltration against postoperative ileus (POI). In this study, we orally administered individual herbal components of DKT to mice four times before and after intestinal manipulation (IM). The anti-inflammatory action of each crude drug was evaluated by histochemical analysis of relevant molecules. The results showed that treatment with all herbal components of DKT significantly inhibits neutrophil infiltration. This inhibition of neutrophil infiltration by ZPR was significantly reduced in 5-hydroxytryptamine receptor 4 (5-HT4R) knockout (KO) mice but not in alpha-7 nicotinic acetylcholine receptor (α7nAChR) KO mice. Also, transient receptor potential ankyrin 1 (TRPA1) and muscarinic acetylcholine receptor (mAChR) antagonists partly and significantly inhibited the amelioration of neutrophil infiltration by ZPR. Therefore, DKT-induced anti-inflammatory action, mediated by inhibition of neutrophil infiltration in POI, depends, in part, on the effects of ZPR. ZPR activates TRPA1 channels, possibly in enterochromaffin (EC) cells, to release 5-HT. This 5-HT stimulates 5-HT4R in the myenteric plexus neurons to release acetylcholine, which, in turn, activates mAChR to inhibit inflammation in POI.
Collapse
Affiliation(s)
- Mari Endo
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Japan.
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Japan.
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Japan
| | - Tetsuro Oikawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Japan
| | - Hiroshi Odaguchi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Japan
| | - Toshihiko Hanawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Japan; Department of Oriental Medicine Research, Research and Development Center for Medical Education, Kitasato University School of Medicine, Japan; Department of Oriental Medicine, Doctoral Program of Medical Science, Kitasato University Graduate School, Japan
| |
Collapse
|
19
|
Matsumoto K, Kawanaka H, Hori M, Kusamori K, Utsumi D, Tsukahara T, Amagase K, Horie S, Yamamoto A, Ozaki H, Mori Y, Kato S. Role of transient receptor potential melastatin 2 in surgical inflammation and dysmotility in a mouse model of postoperative ileus. Am J Physiol Gastrointest Liver Physiol 2018; 315:G104-G116. [PMID: 29565641 DOI: 10.1152/ajpgi.00305.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this study, we investigated the role of transient receptor potential melastatin 2 (TRPM2), a nonselective cation channel abundantly expressed in inflammatory cells such as macrophages, in the development of postoperative ileus, a complication of abdominal surgery characterized by gastrointestinal dysmotility. In wild-type mice, we found that intestinal manipulation, a maneuver that elicits symptoms typical of postoperative ileus, delays the transit of fluorescein-labeled dextran, promotes the infiltration of CD68+ macrophages, Ly6B.2+ neutrophils, and MPO+ cells into intestinal muscles, boosts expression of IL-1β, IL-6, TNF-α, iNOS, and CXCL2 in intestinal muscles and peritoneal macrophages, enhances phosphorylation of ERK and p38 MAPK in intestinal muscles, and amplifies IL-1β, IL-6, TNF-α, iNOS, and CXCL2 expression in resident and thioglycolate-elicited peritoneal macrophages following exposure to lipopolysaccharide. Remarkably, TRPM2 deficiency completely blocks or diminishes these effects. Indeed, intestinal manipulation appears to activate TRPM2 in resident muscularis macrophages and elicits release of inflammatory cytokines and chemokines, which, in turn, promote infiltration of macrophages and neutrophils into the muscle, ultimately resulting in dysmotility. NEW & NOTEWORTHY Activation of transient receptor potential melastatin 2 (TRPM2) releases inflammatory cytokines and chemokines, which, in turn, promote the infiltration of inflammatory cells and macrophages into intestinal muscles, ultimately resulting in dysmotility. Thus TRPM2 is a promising target in treating dysmotility due to postoperative ileus, a complication of abdominal surgery.
Collapse
Affiliation(s)
- Kenjiro Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Hiroki Kawanaka
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Kosuke Kusamori
- Division of Clinical Pharmaceutical Sciences, Department of Biopharmaceutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Daichi Utsumi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Takuya Tsukahara
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Kikuko Amagase
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Syunji Horie
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Josai International University , Chiba , Japan
| | - Akira Yamamoto
- Division of Clinical Pharmaceutical Sciences, Department of Biopharmaceutics, Kyoto Pharmaceutical University , Kyoto , Japan
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Kyoto , Japan
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University , Kyoto , Japan
| |
Collapse
|
20
|
Ileocecal Anastomosis Type Significantly Influences Long-Term Functional Status, Quality of Life, and Healthcare Utilization in Postoperative Crohn's Disease Patients Independent of Inflammation Recurrence. Am J Gastroenterol 2018; 113:576-583. [PMID: 29610509 DOI: 10.1038/ajg.2018.13] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Anastomotic reconstruction following intestinal resection in Crohn's disease (CD) may employ side-to-side anastomosis (STSA; anti-peristaltic orientation) or end-to-end anastomosis (ETEA). Our aim was to determine the impact of these two anastomotic techniques on long-term clinical status in postoperative CD patients. METHODS We performed a comparative effectiveness study of prospectively collected observational data from consented CD patients undergoing their first or second ileocolonic bowel resection and re-anastomosis between 2008 and 2012, in order to assess the association between anastomosis type and 2-year postoperative quality of life (QoL), healthcare utilization, disease clinical or endoscopic recurrence, use of medications, and need for repeat resection. RESULTS One hundred and twenty eight postoperative CD patients (60 STSA and 68 ETEA) were evaluated. At 2 years postoperatively, STSA patients had higher rates of emergency department visits (33.3% vs. 14.7%; P=0.01), hospitalizations (30% vs. 11.8%; P=0.01), and abdominal computed tomography scans (50% vs. 13.2%; P<0.001) with lower QoL (mean short inflammatory bowel disease questionnaire 47.9 vs. 53.4; P=0.007). There was no difference among the two groups in the 30 day surgical complications and 2-year patterns of disease activity, CD medication requirement, endoscopic recurrence, and need for new surgical management (all P > 0.05). CONCLUSIONS At 2 years postoperatively, CD patients with ETEA demonstrated better QoL and less healthcare utilization compared with STSA, despite having similar patterns of disease recurrence and CD treatment. These findings suggest that surgical reconstruction of the bowel as an intact tube (ETEA) contribute to improved functional and clinical status in patients with CD.
Collapse
|
21
|
De Schepper S, Stakenborg N, Matteoli G, Verheijden S, Boeckxstaens GE. Muscularis macrophages: Key players in intestinal homeostasis and disease. Cell Immunol 2017; 330:142-150. [PMID: 29291892 PMCID: PMC6108422 DOI: 10.1016/j.cellimm.2017.12.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022]
Abstract
Muscularis macrophages densily colonize the outermost layer of the gastrointestinal tract. Muscularis macrophages communicate with enteric neurons in a bidirectional matter. Muscularis macrophages are tissue-protective but can contribute to disease. Current challenges are to decipher therapeutic potentials of muscularis macrophages.
Macrophages residing in the muscularis externa of the gastrointestinal tract are highly specialized cells that are essential for tissue homeostasis during steady-state conditions as well as during disease. They are characterized by their unique protective functional phenotype that is undoubtedly a consequence of the reciprocal interaction with their environment, including the enteric nervous system. This muscularis macrophage-neuron interaction dictates intestinal motility and promotes tissue-protection during injury and infection, but can also contribute to tissue damage in gastrointestinal disorders such as post-operative ileus and gastroparesis. Although the importance of muscularis macrophages is clearly recognized, different aspects of these cells remain largely unexplored such their origin, longevity and instructive signals that determine their function and phenotype. In this review, we will discuss the phenotype, functions and origin of muscularis macrophages during steady-state and disease conditions. We will highlight the bidirectional crosstalk with neurons and potential therapeutic strategies that target and manipulate muscularis macrophages to restore their protective signature as a treatment for disease.
Collapse
Affiliation(s)
- Sebastiaan De Schepper
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Nathalie Stakenborg
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Laboratory for Mucosal Immunology, University of Leuven, Leuven, Belgium.
| | - Simon Verheijden
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Guy E Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| |
Collapse
|
22
|
Endo M, Hori M, Mihara T, Ozaki H, Oikawa T, Odaguchi H, Hanawa T. Zingiberis Siccatum Rhizoma, the active component of the Kampo formula Daikenchuto, induces anti-inflammatory actions through α7 nicotinic acetylcholine receptor activation. Neurogastroenterol Motil 2017; 29. [PMID: 28656709 DOI: 10.1111/nmo.13139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/23/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND We previously reported that Daikenchuto (DKT), a gastrointestinal prokinetic Japanese herbal (Kampo) medicine used for the treatment of postoperative ileus (POI), has characteristic potent anti-inflammatory activity. This effect may be partly mediated by the activation of α7 nicotinic acetylcholine receptor (nAChR). In this study, we identified the specific herbs in DKT that induce anti-inflammatory action. METHODS The herbal components of DKT were individually administered orally to each mouse four times before and after intestinal manipulation (IM) was carried out on the distal ileum. The anti-inflammatory activity of each crude drug was subsequently evaluated using immunohistochemical analyses of relevant molecules. KEY RESULTS Treatment with Zingiberis Siccatum Rhizoma (ZSR) but not the other components inhibited the infiltration of cluster of differentiation 68 (CD68)-positive macrophages as effectively as DKT treatment. Selective α7nAChR antagonists, such as methyllycaconitine citrate, or transient receptor potential ankyrin 1 (TRPA1) antagonists, such as HC-030031, significantly inhibited the amelioration of macrophage infiltration by ZSR. The inhibition of macrophage infiltration by ZSR was abolished in both α7nAChR and 5-hydroxytryptamine 4 receptor (5-HT4 R) knockout mice. CONCLUSIONS & INFERENCES Daikenchuto-induced anti-inflammatory activity, which was mediated by inhibiting macrophage infiltration in POI, is dependent on the effects of ZSR. Zingiberis Siccatum Rhizoma activates TRPA1 channels possibly in enterochromaffin (EC) cells to release 5-HT, which stimulates 5-HT4 R in the myenteric plexus neurons to release ACh, which in turn activates α7nAChR on macrophages to inhibit inflammation in POI.
Collapse
Affiliation(s)
- M Endo
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - M Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - T Mihara
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - H Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - T Oikawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - H Odaguchi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - T Hanawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Tokyo, Japan.,Department of Oriental Medicine Research, Research and Development Center for Medical Education, Kitasato University School of Medicine, Tokyo, Japan.,Department of Oriental Medicine, Doctoral Program of Medical Science, Kitasato University Graduate School, Tokyo, Japan
| |
Collapse
|
23
|
Ziegelhoeffer T, Heil M, Fischer S, Fernández B, Schaper W, Preissner KT, Deindl E, Pagel JI. Role of early growth response 1 in arteriogenesis: Impact on vascular cell proliferation and leukocyte recruitment in vivo. Thromb Haemost 2017; 107:562-74. [DOI: 10.1160/th11-07-0490] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 12/13/2011] [Indexed: 02/07/2023]
Abstract
SummaryBased on previous findings that early growth response 1 (Egr-1) participates in leukocyte recruitment and cell proliferation in vitro, this study was designed to investigate its mode of action during arteriogenesis in vivo. In a model of peripheral arteriogenesis, Egr-1 was significantly upregulated in growing collaterals of wild-type (WT) mice, both on mRNA and protein level. Egr-1−/− mice demonstrated delayed arteriogenesis after femoral artery ligation. They further showed increased levels of monocytes and granulocytes in the circulation, but reduced levels in adductor muscles under baseline conditions. After femoral artery ligation, elevated numbers of macrophages were detected in the perivascular zone of collaterals in Egr-1−/− mice and mRNA of leukocyte recruitment mediators was upregulated. Other Egr family members (Egr-2 to -4) were significantly upregulated only in Egr-1−/− mice, suggesting a mechanism of counterbalancing Egr-1 deficiency. Moreover, splicing factor-1, downregulated in WT mice after femoral artery ligation in the process of increased vascular cell proliferation, was upregulated in Egr-1−/− mice. αSM-actin on the other hand, significantly downregulated in WT mice, showed no differential expression in Egr-1−/− mice. While cell cycle regulator cyclin E and cdc20 were upregulated in Egr-1−/− mice, cyclin D1 expression decreased below the detection limit in collaterals, and the proliferation marker ki67 was not differentially expressed. In conclusion, compensation for deficiency in Egr-1 function in leukocyte recruitment can presumably be mediated by other transcription factors; however, Egr-1 is indispensable for effective vascular cell cycle progression in arteriogenesis.
Collapse
|
24
|
Stakenborg N, Gomez-Pinilla PJ, Boeckxstaens GE. Postoperative Ileus: Pathophysiology, Current Therapeutic Approaches. Handb Exp Pharmacol 2017; 239:39-57. [PMID: 27999957 DOI: 10.1007/164_2016_108] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Postoperative ileus, which develops after each abdominal surgical procedure, is an iatrogenic disorder characterized by a transient inhibition of gastrointestinal motility. Its pathophysiology is complex involving pharmacological (opioids, anesthetics), neural, and immune-mediated mechanisms. The early neural phase, triggered by activation of afferent nerves during the surgical procedure, is short lasting compared to the later inflammatory phase. The latter starts after 3-6 h and lasts several days, making it a more interesting target for treatment. Insight into the triggers and immune cells involved is of great importance for the development of new therapeutic strategies. In this chapter, the pathogenesis and the current therapeutic approaches to treat postoperative ileus are discussed.
Collapse
Affiliation(s)
- N Stakenborg
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - P J Gomez-Pinilla
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - G E Boeckxstaens
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium. .,Division of Gastroenterology and Hepatology, University Hospital Leuven, Herestraat 49, Leuven, 3000, Belgium.
| |
Collapse
|
25
|
Fang JF, Fang JQ, Shao XM, Du JY, Liang Y, Wang W, Liu Z. Electroacupuncture treatment partly promotes the recovery time of postoperative ileus by activating the vagus nerve but not regulating local inflammation. Sci Rep 2017; 7:39801. [PMID: 28051128 PMCID: PMC5209726 DOI: 10.1038/srep39801] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
Postoperative ileus (POI) after abdominal surgery significantly lowers the life quality of patients and increase hospital costs. However, few treatment strategies have successfully shortened the duration of POI. Electroacupuncture (EA) is a modern way of administering acupuncture and widely used in various gastrointestinal (GI) diseases in the world. Here, we studied the effect of EA on POI and its underlying mechanisms. Intestinal manipulation resulted in significant delays of GI transit, colonic transit and gastric emptying. Surgery also up-regulated c-fos in nucleus of the solitary tract (NTS) and induced inflammation response in the small intestine. Further, operation and inhale anesthesia inhibited NTS neuron excitation duration for the whole observation time. EA administered at ST36 indeed shortened the recovery time of GI and colonic transit, and significantly increased the gastric emptying. EA also significantly activated the NTS neurons after operation. However, there was no anti-inflammation effect of EA during the whole experiment. Finally, atropine blocked the regulatory effect of EA on GI function, when it was injected after surgery, but not before surgery. Thus, the regulatory effect of EA on POI was mainly mediated by exciting NTS neurons to improve the GI tract transit function but not by activating cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Jun-Fan Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian-Qiao Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao-Mei Shao
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun-Ying Du
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Liang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen Wang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhe Liu
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
26
|
Farro G, Gomez-Pinilla PJ, Di Giovangiulio M, Stakenborg N, Auteri M, Thijs T, Depoortere I, Matteoli G, Boeckxstaens GE. Smooth muscle and neural dysfunction contribute to different phases of murine postoperative ileus. Neurogastroenterol Motil 2016; 28:934-47. [PMID: 26891411 DOI: 10.1111/nmo.12796] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/15/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Postoperative ileus (POI) is characterized by a transient inhibition of gastrointestinal (GI) motility after abdominal surgery mediated by the inflammation of the muscularis externa (ME). The aim of this study was to identify alterations in the enteric nervous system that may contribute to the pathogenesis of POI. METHODS Gastrointestinal transit, contractility of isolated smooth muscle strips and inflammatory parameters were evaluated at different time points (1.5 h to 10 days) after intestinal manipulation (IM) in mice. Immune-labeling was used to visualize changes in myenteric neurons. KEY RESULTS Intestinal manipulation resulted in an immediate inhibition of GI transit recovering between 24 h and 5 days. In vitro contractility to K(+) (60 mM) or carbachol (10(-9) to 10(-4) M) was biphasically suppressed over 24 h after IM (with transient recovery at 6 h). The first phase of impaired myogenic contractility was associated with increased expression of TNF-α, IL-6 and IL-1α. After 24 h, we identified a significant reduction in electrical field stimulation-evoked contractions and relaxations, lasting up to 10 days after IM. This was associated with a reduced expression of chat and nos1 genes. CONCLUSIONS & INFERENCES Intestinal manipulation induces two waves of smooth muscle inhibition, most likely mediated by inflammatory cytokines, lasting up to 3 days after IM. Further, we here identify a late third phase (>24 h) characterized by impaired cholinergic and nitrergic neurotransmission persisting after recovery of muscle contractility. These findings illustrate that POI results from inflammation-mediated impaired smooth muscle contraction, but also involves a long-lasting impact of IM on the enteric nervous system.
Collapse
Affiliation(s)
- G Farro
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - P J Gomez-Pinilla
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - M Di Giovangiulio
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - N Stakenborg
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - M Auteri
- Division of Physiology, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - T Thijs
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - I Depoortere
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Matteoli
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Ryu WI, Lee H, Kim JH, Bae HC, Ryu HJ, Son SW. IL-33 induces Egr-1-dependent TSLP expression via the MAPK pathways in human keratinocytes. Exp Dermatol 2015; 24:857-63. [DOI: 10.1111/exd.12788] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Woo-In Ryu
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Hana Lee
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Jin Hee Kim
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Hyun Cheol Bae
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Hwa Jung Ryu
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Sang Wook Son
- Department of Dermatology; Division of Brain Korea 21 Project for Biomedical Science; Korea University College of Medicine; Seoul Korea
| |
Collapse
|
28
|
Fraguas S, Barberán S, Iglesias M, Rodríguez-Esteban G, Cebrià F. egr-4, a target of EGFR signaling, is required for the formation of the brain primordia and head regeneration in planarians. Development 2014; 141:1835-47. [PMID: 24700819 DOI: 10.1242/dev.101345] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
During the regeneration of freshwater planarians, polarity and patterning programs play essential roles in determining whether a head or a tail regenerates at anterior or posterior-facing wounds. This decision is made very soon after amputation. The pivotal role of the Wnt/β-catenin and Hh signaling pathways in re-establishing anterior-posterior (AP) polarity has been well documented. However, the mechanisms that control the growth and differentiation of the blastema in accordance with its AP identity are less well understood. Previous studies have described a role of Smed-egfr-3, a planarian epidermal growth factor receptor, in blastema growth and differentiation. Here, we identify Smed-egr-4, a zinc-finger transcription factor belonging to the early growth response gene family, as a putative downstream target of Smed-egfr-3. Smed-egr-4 is mainly expressed in the central nervous system and its silencing inhibits anterior regeneration without affecting the regeneration of posterior regions. Single and combinatorial RNA interference to target different elements of the Wnt/β-catenin pathway, together with expression analysis of brain- and anterior-specific markers, revealed that Smed-egr-4: (1) is expressed in two phases - an early Smed-egfr-3-independent phase and a late Smed-egfr-3-dependent phase; (2) is necessary for the differentiation of the brain primordia in the early stages of regeneration; and (3) that it appears to antagonize the activity of the Wnt/β-catenin pathway to allow head regeneration. These results suggest that a conserved EGFR/egr pathway plays an important role in cell differentiation during planarian regeneration and indicate an association between early brain differentiation and the proper progression of head regeneration.
Collapse
Affiliation(s)
- Susanna Fraguas
- Departament de Genètica de la Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Avenida Diagonal 643, Edifici Prevosti planta 1, Barcelona 08028, Spain
| | | | | | | | | |
Collapse
|
29
|
Attenuation of Inflammatory Response and Reduction in Infarct Size by Postconditioning Are Associated With Downregulation of Early Growth Response 1 During Reperfusion in Rat Heart. Shock 2014; 41:346-54. [DOI: 10.1097/shk.0000000000000112] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
30
|
Endo M, Hori M, Ozaki H, Oikawa T, Hanawa T. Daikenchuto, a traditional Japanese herbal medicine, ameliorates postoperative ileus by anti-inflammatory action through nicotinic acetylcholine receptors. J Gastroenterol 2014; 49:1026-39. [PMID: 23846546 PMCID: PMC4048467 DOI: 10.1007/s00535-013-0854-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/15/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Daikenchuto (DKT), a gastrointestinal prokinetic Japanese herbal medicine, is prescribed for patients with postoperative ileus (POI) and adhesive bowel obstruction following abdominal surgery. Several mechanisms for the amelioration of POI by DKT have been suggested; however, it has remained unclear whether DKT shows anti-inflammatory effects in POI. In the present study, we investigated the effects of DKT in a mouse POI model and attempted to clarify the detailed mechanisms of action. METHOD Intestinal manipulation (IM) was applied to the distal ileum of mice. DKT was administered orally to the animals 4 times before and after IM. Gastrointestinal transit in vivo, leukocyte infiltration, cytokine mRNA expression and gastrointestinal motility were analyzed. We also investigated the effects of the α7nAChR antagonist methyllycaconitine citrate (MLA) on the DKT-mediated ameliorative action against POI, and we studied the effects of DKT on inflammatory activity in α7nAChR knockout mice. RESULTS DKT treatment led to recovery of the delayed intestinal transit induced by IM. DKT significantly inhibited the infiltration of neutrophils and CD68-positive macrophages, and inhibited mRNA expressions of TNF-α and MCP-1. MLA significantly reduced the anti-inflammatory action of DKT, and the amelioration of macrophage infiltration by DKT was partially suppressed in α7nAChR knockout mice. CONCLUSIONS In conclusion, in addition to the gastrointestinal prokinetic action, DKT serves as a novel therapeutic agent for POI characterized by its anti-inflammatory potency. The DKT-induced anti-inflammatory activity may be partly mediated by activation of α7nAChR.
Collapse
Affiliation(s)
- Mari Endo
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8642 Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657 Japan
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657 Japan
| | - Tetsuro Oikawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8642 Japan
| | - Toshihiko Hanawa
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8642 Japan ,Department of Oriental Medicine, Doctoral Program of Medical Science, Kitasato University Graduate School, 1-15-1 kitasato, Sagamihara-shi, Kanagawa, 228-8555 Japan
| |
Collapse
|
31
|
Endo M, Hori M, Ozaki H, Oikawa T, Hanawa T. Rikkunshito, a Kampo Medicine, Ameliorates Post-operative Ileus by Anti-inflammatory Action. J Pharmacol Sci 2014; 124:374-85. [DOI: 10.1254/jphs.13182fp] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
32
|
van Bree SHW, Gomez-Pinilla PJ, van de Bovenkamp FS, Di Giovangiulio M, Farro G, Nemethova A, Cailotto C, de Jonge WJ, Lee K, Ramirez-Molina C, Lugo D, Skynner MJ, Boeckxstaens GEE, Matteoli G. Inhibition of spleen tyrosine kinase as treatment of postoperative ileus. Gut 2013; 62:1581-90. [PMID: 23242119 DOI: 10.1136/gutjnl-2012-302615] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Intestinal inflammation resulting from manipulation-induced mast cell activation is a crucial mechanism in the pathophysiology of postoperative ileus (POI). Recently it has been shown that spleen tyrosine kinase (Syk) is involved in mast cell degranulation. Therefore, we have evaluated the effect of the Syk-inhibitor GSK compound 143 (GSK143) as potential treatment to shorten POI. DESIGN In vivo: in a mouse model of POI, the effect of the Syk inhibitor (GSK143) was evaluated on gastrointestinal transit, muscular inflammation and cytokine production. In vitro: the effect of GSK143 and doxantrazole were evaluated on cultured peritoneal mast cells (PMCs) and bone marrow derived macrophages. RESULTS In vivo: intestinal manipulation resulted in a delay in gastrointestinal transit at t=24 h (Geometric Center (GC): 4.4 ± 0.3). Doxantrazole and GSK143 significantly increased gastrointestinal transit (GC doxantrazole (10 mg/kg): 7.2 ± 0.7; GSK143 (1 mg/kg): 7.6 ± 0.6), reduced inflammation and prevented recruitment of immune cells in the intestinal muscularis. In vitro: in PMCs, substance P (0-90 μM) and trinitrophenyl (0-4 μg/ml) induced a concentration-dependent release of β-hexosaminidase. Pretreatment with doxantrazole and GSK143 (0.03-10 μM) concentration dependently blocked substance P and trinitrophenyl induced β-hexosaminidase release. In addition, GSK143 was able to reduce cytokine expression in endotoxin-treated bone marrow derived macrophages in a concentration-dependent manner. CONCLUSIONS The Syk inhibitor GSK143 reduces macrophage activation and mast cell degranulation in vitro. In addition, it inhibits manipulation-induced intestinal muscular inflammation and restores intestinal transit in mice. These findings suggest that Syk inhibition may be a new tool to shorten POI.
Collapse
Affiliation(s)
- Sjoerd H W van Bree
- Department of Gastroenterology and Hepatology, Tytgat Institute of Liver and Intestinal Research, Academic Medical Center, , Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
van Bree SHW, Cailotto C, Di Giovangiulio M, Jansen E, van der Vliet J, Costes L, Depoortere I, Gomez-Pinilla PJ, Matteoli G, Boeckxstaens GEE. Systemic inflammation with enhanced brain activation contributes to more severe delay in postoperative ileus. Neurogastroenterol Motil 2013; 25:e540-9. [PMID: 23711101 DOI: 10.1111/nmo.12157] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/26/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND The severity of postoperative ileus (POI) has been reported to result from decreased contractility of the muscularis inversely related to the number of infiltrating leukocytes. However, we previously observed that the severity of POI is independent of the number of infiltrating leukocytes, indicating that different mechanisms must be involved. Here, we hypothesize that the degree of tissue damage in response to intestinal handling determines the upregulation of local cytokine production and correlates with the severity of POI. METHODS Intestinal transit, the inflammatory response, I-FABP (marker for tissue damage) levels and brain activation were determined after different intensities of intestinal handling. KEY RESULTS Intense handling induced a more pronounced ileus compared with gentle intestinal manipulation (IM). No difference in leukocytic infiltrates in the handled and non-handled parts of the gut was observed between the two intensities of intestinal handling. However, intense handling resulted in significantly more tissue damage and was accompanied by a systemic inflammation with increased plasma levels of pro-inflammatory cytokines. In addition, intense but not gentle handling triggered enhanced c-Fos expression in the nucleus of the solitary tract (NTS) and area postrema (AP). In patients, plasma levels of I-FABP and inflammatory cytokines were significantly higher after open compared with laparoscopic surgery, and were associated with more severe POI. CONCLUSIONS & INFERENCES Not the influx of leukocytes, rather the manipulation-induced damage and subsequent inflammatory response determine the severity of POI. The release of tissue damage mediators and pro-inflammatory cytokines into the systemic circulation most likely contribute to the impaired motility of non-manipulated intestine.
Collapse
Affiliation(s)
- S H W van Bree
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
van Bree SH, Nemethova A, van Bovenkamp FS, Gomez-Pinilla P, Elbers L, Di Giovangiulio M, Matteoli G, van Vliet J, Cailotto C, Tanck MW, Boeckxstaens GE. Novel method for studying postoperative ileus in mice. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2012; 4:219-227. [PMID: 23320135 PMCID: PMC3544220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Accepted: 11/15/2012] [Indexed: 06/01/2023]
Abstract
INTRODUCTION Postoperative ileus (POI) is characterized by a transient inhibition of coordinated motility of the gastrointestinal (GI) tract after abdominal surgery and leads to increased morbidity and prolonged hospitalization. Currently, intestinal manipulation of the intestine is widely used as a preclinical model of POI. The technique used to manipulate the intestine is however highly variable and difficult to standardize, leading to large variations and inconsistent findings between different investigators. Therefore, we developed a device by which a fixed and adjustable pressure can be applied during intestinal manipulation. METHODS The standardized pressure manipulation method was developed using the purpose-designed device. First, the effect of graded manipulation was examined on postoperative GI transit. Next, this new technique was compared to the conventional manipulation technique used in previous studies. GI transit was measured by evaluating the intestinal distribution of orally gavaged fluorescein isothiocyanate (FITC)-labeled dextran. Infiltration of myeloperoxidase positive cells and cytokine production (ELISA) in the muscularis externa of the intestine were assessed. RESULTS Increasing pressures resulted in a graded reduction of intestinal transit and was associated with intestinal inflammation as demonstrated by influx of leukocytes and increased levels of IL-6, IL-1β and MCP-1 compared to control mice. With an applied pressure of 9 grams a similar delay in intestinal transit could be obtained with a smaller standard deviation, leading to a reduced intra-individual variation. CONCLUSIONS This method provides a reproducible model with small variation to study the pathophysiology of POI and to evaluate new anti-inflammatory strategies.
Collapse
Affiliation(s)
- Sjoerd Hw van Bree
- Tytgat institute of Liver and Intestinal Research, Department of Gastroenterology & Hepatology, Academic Medical Center Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang J, Xie S, Ma W, Teng Y, Tian Y, Huang X, Zhang Y. A newly identified microRNA, mmu-miR-7578, functions as a negative regulator on inflammatory cytokines tumor necrosis factor-α and interleukin-6 via targeting Egr1 in vivo. J Biol Chem 2012. [PMID: 23184950 DOI: 10.1074/jbc.m112.351197] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Appropriate innate immune responses are required to protect an organism against foreign pathogens, and the immune response must be tightly controlled. Here, we report a new microRNA (miRNA) identified from a small RNA library from the epididymis, termed miR-7578, that acts as a negative regulator of inflammatory responses. It was abundantly expressed in immune-related organs and induced by lipopolysaccharide in the lung and epididymis, as well as macrophages stimulated with diverse Toll-like receptor ligands, in an NF-κB-dependent manner. mmu-miR-7578 inhibited the release of pro-inflammatory cytokines, including TNFα and IL6, by regulating its target gene Egr1, which encodes a transcription factor that activates TNFα and NF-κB expression. Transgenic mice overexpressing mmu-miR-7578 displayed higher resistance to endotoxin shock and lower plasma levels of TNFα and IL6, indicating that this miRNA acted as a negative molecule of immune response. In sum, we report a previously uncharacterized LPS-responsive miRNA that controls inflammatory response in a feedback loop by fine-tuning a key transcription factor in vivo.
Collapse
Affiliation(s)
- Jinsong Zhang
- Shanghai Key Laboratory of Molecular Andrology, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Yu W, Lin Z, Hegarty JP, Chen X, Kelly AA, Wang Y, Poritz LS, Koltun WA. Genes differentially regulated by NKX2-3 in B cells between ulcerative colitis and Crohn's disease patients and possible involvement of EGR1. Inflammation 2012; 35:889-99. [PMID: 21968973 DOI: 10.1007/s10753-011-9390-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD) are two related yet different forms of chronic intestinal inflammation. We investigated the genes regulated by NKX2-3 in B cells from a UC patient by cDNA microarray and compared the results to those genes regulated by NKX2-3 in B cells from a CD patient. Genes regulated by NKX2-3 in B cells from UC were mainly involved in cell growth, inflammation, and immune response. Among the genes regulated by NKX2-3 in both UC and CD, expression of 145 genes was similarly altered and 34 genes was differentially affected by NKX2-3 knockdown. EGR1 was up-regulated in NKX2-3 knockdown B cells from UC while down-regulated in NKX2-3 knockdown B cells from CD. mRNA expressions of NKX2-3 and EGR1 were increased in diseased intestinal tissues from 19 CD patients. NKX2-3 may play different roles in UC and CD pathogenesis by differential regulation of EGR1.
Collapse
Affiliation(s)
- Wei Yu
- Department of Colon and Rectal Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Fan YY, Ye GH, Lin KZ, Yu LS, Wu SZ, Dong MW, Han JG, Feng XP, Li XB. Time-dependent expression and distribution of Egr-1 during skeletal muscle wound healing in rats. J Mol Histol 2012; 44:75-81. [PMID: 22918836 DOI: 10.1007/s10735-012-9445-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 08/14/2012] [Indexed: 12/26/2022]
Abstract
Recent studies have shown that early growth response factor-1 (Egr-1) plays an important role in regulation of inflammation and tissue repair, but little is known about its expression after trauma to skeletal muscles. A preliminary study on time-dependent expression and distribution of Egr-1 was performed by immunohistochemistry, immunofluorescence and Western blotting during skeletal muscle wound healing in rats. An animal model of skeletal muscle contusion was established in 45 Sprague-Dawley male rats. Samples were taken at 6 h, 12 h, 1 day, 3 days, 5 days, 7 days, 10 days, 14 days and 21 days post-injury, respectively (5 rats in each posttraumatic interval). 5 rats were employed as control. In the uninjured controls, Egr-1 positive staining was observed in the sarcoplasm and nuclei of normal myofibers. In wounded specimens, a small number of polymorphonuclear cells (PMNs), a number of mononuclear cells (MNCs), fibroblastic cells (FBCs) and regenerated multinucleated myotubes showed positive reaction for Egr-1 in contused zones. By morphometric analysis, an increase in Egr-1 expression was verified at inflammatory phase after contusion, which reached a peak in the regenerated phase overlapping with the fibrotic phase during skeletal muscle wound healing. The expression tendency was further confirmed by Western blotting assay. By immunofluorescent staining for co-localization, the Egr-1-positive MNCs and FBCs in wounds were identified as macrophages and myofibroblasts. The results demonstrate that the expression of Egr-1 is up-regulated and temporally distributed in certain cell types after trauma to skeletal muscles, which may be closely involved in inflammatory response, fibrotic repair and muscle regeneration during skeletal muscle wound healing.
Collapse
Affiliation(s)
- Yan-Yan Fan
- Department of Forensic Medicine, School of Basic Medical Science, Wenzhou Medical College, Higher Education District, Wenzhou, Zhejiang Province, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Buchholz BM, Chanthaphavong RS, Billiar TR, Bauer AJ. Role of interleukin-6 in hemopoietic and non-hemopoietic synergy mediating TLR4-triggered late murine ileus and endotoxic shock. Neurogastroenterol Motil 2012; 24:658-69, e294. [PMID: 22489868 PMCID: PMC3378783 DOI: 10.1111/j.1365-2982.2012.01914.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Early murine endotoxin-induced ileus at 6 h is exclusively mediated by non-hemopoietic TLR4/MyD88 signaling despite molecular activation of hemopoietic cells which included a significant IL-6 mRNA induction. Our objective was to define the role of hemopoietic cells in LPS/TLR4-triggered ileus and inflammation over time, and identify mechanisms of ileus. METHODS CSF-1(-/-) , TLR4 non-chimera and TLR4 chimera mice were single-shot intraperitoneal injected with ultrapure lipopolysaccharide (UP-LPS) and studied up to 4 days. Subgroups of TLR4(WT) mice were additionally intravenously injected with exogenous recombinant IL-6 (rmIL-6) or murine soluble IL-6 receptor blocking antibody (anti-sIL-6R mAB). KEY RESULTS Hemopoietic TLR4 signaling independently mediated UP-LPS-induced ileus at 24 h, but chemotactic muscularis neutrophil extravasation was not causatively involved and mice lacking CSF-1-dependent macrophages died prematurely. Synergy of hemopoietic and non-hemopoietic cells determined ileus severity and mortality which correlated with synergistic cell lineage specific transcription of inflammatory mediators like IL-6 within the intestinal muscularis. Circulating IL-6 levels were LPS dose dependent, but exogenous rmIL-6 did not spark off a self-perpetuating inflammatory response triggering ileus. Sustained therapeutic inhibition of functional IL-6 signaling efficiently ameliorated late ileus while preemptive antibody-mediated IL-6R blockade was marginally effective in mitigating ileus. However, IL-6R blockade did not prevent endotoxin-associated mortality nor did it alter circulating IL-6 levels. CONCLUSIONS & INFERENCES A time-delayed bone marrow-driven mechanism of murine endotoxin-induced ileus exists, and hemopoietic cells synergize with non-hemopoietic cells thereby prolonging ileus and fueling intestinal inflammation. Importantly, IL-6 signaling via IL-6R/gp130 drives late ileus, yet it did not regulate mortality in endotoxic shock.
Collapse
Affiliation(s)
- Bettina M. Buchholz
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA,Department of Surgery, University of Bonn, Bonn, Germany
| | | | | | - Anthony J. Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
39
|
Skorokhod A, Bachmann J, Giese NA, Martignoni ME, Krakowski-Roosen H. Real-imaging cDNA-AFLP transcript profiling of pancreatic cancer patients: Egr-1 as a potential key regulator of muscle cachexia. BMC Cancer 2012; 12:265. [PMID: 22721276 PMCID: PMC3465185 DOI: 10.1186/1471-2407-12-265] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 05/24/2012] [Indexed: 01/08/2023] Open
Abstract
Background Cancer cachexia is a progressive wasting syndrome and the most prevalent characteristic of cancer in patients with advanced pancreatic adenocarcinoma. We hypothesize that genes expressed in wasted skeletal muscle of pancreatic cancer patients may determine the initiation and severity of cachexia syndrome. Experimental design We studied gene expression in skeletal muscle biopsies from pancreatic cancer patients with and without cachexia utilizing Real-Imaging cDNA-AFLP-based transcript profiling for genome-wide expression analysis. Results Our approach yielded 183 cachexia-associated genes. Ontology analysis revealed characteristic changes for a number of genes involved in muscle contraction, actin cytoskeleton rearrangement, protein degradation, tissue hypoxia, immediate early response and acute-phase response. Conclusions We demonstrate that Real-Imaging cDNA-AFLP analysis is a robust method for high-throughput gene expression studies of cancer cachexia syndrome in patients with pancreatic cancer. According to quantitative RT-PCR validation, the expression levels of genes encoding the acute-phase proteins α-antitrypsin and fibrinogen α and the immediate early response genes Egr-1 and IER-5 were significantly elevated in the skeletal muscle of wasted patients. By immunohistochemical and Western immunoblotting analysis it was shown, that Egr-1 expression is significantly increased in patients with cachexia and cancer. This provides new evidence that chronic activation of systemic inflammatory response might be a common and unifying factor of muscle cachexia.
Collapse
Affiliation(s)
- Alexander Skorokhod
- Division of Preventive Oncology (G110), National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
40
|
Wehner S, Vilz TO, Stoffels B, Kalff JC. Immune mediators of postoperative ileus. Langenbecks Arch Surg 2012; 397:591-601. [PMID: 22382699 DOI: 10.1007/s00423-012-0915-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 01/24/2012] [Indexed: 02/08/2023]
Abstract
UNLABELLED CLINICAL BACKGROUND: In all patients undergoing abdominal surgery, a transient phase of interruption of bowel motility, named postoperative ileus (POI) occurs. POI is often accepted as an unavoidable "physiological" response and a self-limiting complication after surgery although it has a significant impact on patient morbidity with prolonged hospitalization and increased costs. Annual economic burden has been estimated as much as US $1.47 billion in the USA (Iyer et al. in J Manag Care Pharm 15(6):485-494, 2009). PATHOPHYSIOLOGY The pathophysiology has been elucidated within the last decades, demonstrating that both, neurogenic and inflammatory mechanisms are involved in response to the surgical trauma. It is now generally accepted that POI pathogenesis processes in two phases: a first neurogenic phase is accountable for the immediate postoperative impairment of bowel motility. This is followed by a second immunological phase that can last for days and mainly affects strength and length of POI. More recent findings demonstrate a bidirectional interaction between the nervous and the immune system, and this interaction significantly contributed to our present understanding of POI pathophysiology. Although nerval mechanisms have a significant impact in the early phase of POI, the contribution of the immune system and subsequently its manipulation has risen as the most promising strategy in prevention or treatment of the clinically relevant prolonged form of POI. AIMS The present manuscript will give an update on the inflammatory responses, the involved cell types, and participating immune mediators in POI.
Collapse
Affiliation(s)
- Sven Wehner
- Department of Surgery, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | | | | | | |
Collapse
|
41
|
Hydrogen-enriched preservation protects the isogeneic intestinal graft and amends recipient gastric function during transplantation. Transplantation 2011; 92:985-92. [PMID: 21956195 DOI: 10.1097/tp.0b013e318230159d] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Inhaled hydrogen gas exerts antioxidant and anti-inflammatory effects in rat intestinal transplantation. Here, we investigated whether ex vivo donor organ treatment with dissolved hydrogen would prevent intestinal graft injury. METHODS Isogeneic intestinal transplantation was performed in Lewis rats with vascular flush, luminal preservation, and cold graft storage in nitrogen-bubbled (SITxN2) or hydrogen-bubbled (SITxH2) preservation solution. Lactated Ringer's solution and 3-hr cold ischemia time were used for mechanistic investigations, whereas survival experiments were performed with University of Wisconsin solution and 6-hr cold ischemia time. RESULTS During the early phase of ischemia-reperfusion injury, hydrogen-enriched solution significantly preserved mucosal graft morphology, diminished graft malondialdehyde levels demonstrating substantial reduction potential and blunted proinflammatory molecular responses (early growth response gene [EGR-1], interleukin [IL]-6, IL-1ß, and inducible nitric oxide synthase) within the reperfused intestinal graft muscularis. During the late phase of ischemia-reperfusion injury, circulating IL-6 protein and lactate dehydrogenase levels were significantly ameliorated in SITxH2 animals, which were associated with a favorable functional outcome in in vivo liquid gastrointestinal transit and recipient solid gastric emptying of chrome steel balls, and marked prevention of the posttransplant associated suppression of in vitro muscarinic jejunal contractility. Reflecting improved graft preservation, hydrogen preloading of grafts increased recipient survival rates from 41% to 80%. Anti-inflammatory and antiapoptotic heme oxygenase-1 was significantly upregulated in the hydrogen-treated graft muscularis but not mucosa before reperfusion. CONCLUSIONS Graft preloading with hydrogen demonstrated superior morphologic and functional graft protection in rodent intestinal transplantation, ultimately facilitating recipient survival. Antioxidant capacity and muscularis heme oxygenase-1 upregulation are possible protective mechanisms.
Collapse
|
42
|
Tsuchida Y, Hatao F, Fujisawa M, Murata T, Kaminishi M, Seto Y, Hori M, Ozaki H. Neuronal stimulation with 5-hydroxytryptamine 4 receptor induces anti-inflammatory actions via α7nACh receptors on muscularis macrophages associated with postoperative ileus. Gut 2011; 60:638-47. [PMID: 21115544 PMCID: PMC3071096 DOI: 10.1136/gut.2010.227546] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The main symptom of postoperative ileus (POI) is an intestinal motility disorder in which monocytes/macrophages and neutrophils play crucial roles. Prokinetic 5-hydroxytryptamine 4 receptor (5-HT₄R) agonists and dopamine receptor antagonists are potential therapeutic agents for directly ameliorating the motility disorder associated with POI. AIM To determine the effects of the 5-HT₄R agonists mosapride citrate (MOS) and CJ-033466 on intestinal smooth muscle contractility relative to immune reactions after POI. METHODS Intestinal manipulation (IM) was applied to the rat distal ileum. Both MOS (0.3 and 1 mg/kg, s.c.) and CJ-033466 (1 mg/kg, s.c.) were administered to the animals before and after IM. At 24 h after IM, isolated intestinal smooth muscle contractile activity in vitro, gastrointestinal transit in vivo, inflammatory mediator expression and leucocyte infiltration were measured. RESULTS After IM, ileal circular muscle contractility in vitro and gastrointestinal transit in vivo were reduced and the number of macrophages and neutrophils increased in the inflamed muscle layer, resulting in the induction of inflammatory mediators such as interleukin 1 β (IL-1β), IL-6, tumour necrosis factor α (TNFα), monocyte chemoattractant protein 1 (MCP-1) and inducible nitric oxide synthase (iNOS). Both MOS and CJ-033466 significantly attenuated not only the intestinal motility dysfunction but also the leucocyte infiltration and inflammatory mediator expression after IM. The autonomic ganglionic blocker hexamethonium (1 mg/kg, i.p.) and the α7-nicotinic acetylcholine receptor (α7nAChR) antagonist methyl lycaconitine citrate (0.087 mg/kg, i.p.) blocked MOS-mediated ameliorative actions. Immunohistochemically, α7nAChR is expressed by monocytes/macrophages but not by neutrophils in the inflamed intestine. CONCLUSION Stimulating the 5-HT₄R accelerates acetyl choline (ACh) release from cholinergic myenteric neurons, which subsequently activates α7nAChR on activated monocytes/macrophages to inhibit their inflammatory reactions in the muscle layer. In addition to their gastroprokinetic action, 5-HT₄R agonists might serve as novel therapeutic agents for POI characterised by anti-inflammatory potency.
Collapse
Affiliation(s)
- Yasuaki Tsuchida
- Department of Veterinary Pharmacology, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - Fumihiko Hatao
- Department of Metabolic Care and Gastrointestinal Surgery Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiko Fujisawa
- Department of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Takahisa Murata
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Michio Kaminishi
- Department of Metabolic Care and Gastrointestinal Surgery Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Metabolic Care and Gastrointestinal Surgery Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
43
|
Tsukamoto T, Antonic V, El Hajj II, Stojadinovic A, Binion DG, Izadjoo MJ, Yokota H, Pape HC, Bauer AJ. Novel model of peripheral tissue trauma-induced inflammation and gastrointestinal dysmotility. Neurogastroenterol Motil 2011; 23:379-86, e164. [PMID: 21303433 PMCID: PMC3105173 DOI: 10.1111/j.1365-2982.2011.01675.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Trauma is a leading cause of death and although the gut is recognized as the 'motor' of post-traumatic systemic inflammatory response syndrome and multiple organ failure, studies on the gastrointestinal (GI) tract are few. Our objectives were to create a precisely controllable tissue injury model in which GI motility, systemic inflammation and wound fluid can be analyzed. METHODS A non-narcotic murine trauma model was developed by the subcutaneous dorsal trans-implantation of a devitalized donor syngeneic harvested tissue-bone matrix (TBX), which was precisely adjusted to % total body weight and studied after 21 h. Gastrointestinal transit histograms were plotted after the oral administration of non-digestible FITC-dextran and geometric centers calculated. Organ bath evaluated jejunal circular muscle contractility. Multiplex electrochemiluminescence measurements of serum and TBX wound fluid inflammatory mediators were performed. KEY RESULTS Increasing TBX amounts progressively delayed transit, whereas TBX heat denaturation or decellularization prevented ileus and death. In the TBX(17.5%) model, jejunal muscle contractility was suppressed and a systemic inflammatory response developed as significant serum elevations in IL-6, keratinocyte cytokine and IL-10 compared to sham. In addition, inflammatory responses within the wound fluid showed elevated levels of preformed IL-1β and TNF-α, whereas, 21 h after implantation IL-1β, IL-6 and keratinocyte cytokine were significantly increased in the wound. CONCLUSIONS & INFERENCES A novel donor tissue-bone matrix trauma model was developed that is precisely adjustable and recapitulates important clinical phenomena. The non-narcotic model demonstrated that increasing tissue injury progressively caused ileus, initiated a systemic inflammatory response and developed inflammatory changes within the wound.
Collapse
Affiliation(s)
- Takeshi Tsukamoto
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, USA, Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Vlado Antonic
- Combat Wound Initiative Program, Walter Reed Army Medical Center, Washington D.C., USA
| | - Ihab I. El Hajj
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - David G. Binion
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mina J. Izadjoo
- Division of Wound Biology and Translational Research, Armed Forces Institute of Pathology, Washington, D.C., USA
| | - Hiroyuki Yokota
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Hans Christoph Pape
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anthony J. Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
De Winter BY, De Man JG. Interplay between inflammation, immune system and neuronal pathways: Effect on gastrointestinal motility. World J Gastroenterol 2010; 16:5523-35. [PMID: 21105185 PMCID: PMC2992670 DOI: 10.3748/wjg.v16.i44.5523] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a systemic inflammatory response representing the leading cause of death in critically ill patients, mostly due to multiple organ failure. The gastrointestinal tract plays a pivotal role in the pathogenesis of sepsis-induced multiple organ failure through intestinal barrier dysfunction, bacterial translocation and ileus. In this review we address the role of the gastrointestinal tract, the mediators, cell types and transduction pathways involved, based on experimental data obtained from models of inflammation-induced ileus and (preliminary) clinical data. The complex interplay within the gastrointestinal wall between mast cells, residential macrophages and glial cells on the one hand, and neurons and smooth muscle cells on the other hand, involves intracellular signaling pathways, Toll-like receptors and a plethora of neuroactive substances such as nitric oxide, prostaglandins, cytokines, chemokines, growth factors, tryptases and hormones. Multidirectional signaling between the different components in the gastrointestinal wall, the spinal cord and central nervous system impacts inflammation and its consequences. We propose that novel therapeutic strategies should target inflammation on the one hand and gastrointestinal motility, gastrointestinal sensitivity and even pain signaling on the other hand, for instance by impeding afferent neuronal signaling, by activation of the vagal anti-inflammatory pathway or by the use of pharmacological agents such as ghrelin and ghrelin agonists or drugs interfering with the endocannabinoid system.
Collapse
|
45
|
Gurgui M, Broere R, Kalff JC, van Echten-Deckert G. Dual action of sphingosine 1-phosphate in eliciting proinflammatory responses in primary cultured rat intestinal smooth muscle cells. Cell Signal 2010; 22:1727-33. [DOI: 10.1016/j.cellsig.2010.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 06/23/2010] [Accepted: 06/26/2010] [Indexed: 12/30/2022]
|
46
|
Buchholz BM, Billiar TR, Bauer AJ. Dominant role of the MyD88-dependent signaling pathway in mediating early endotoxin-induced murine ileus. Am J Physiol Gastrointest Liver Physiol 2010; 299:G531-8. [PMID: 20508155 PMCID: PMC2928536 DOI: 10.1152/ajpgi.00060.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
TLR4 ligation by pathogen-associated molecular patterns, such as Gram-negative bacteria-derived LPS, triggers a nonhematopoietic cell-mediated ileus during early endotoxemia. Our objective was to investigate the quantitative contributions of the two downstream signaling pathways of TLR4, namely the adapter proteins myeloid differentiation primary response gene 88 (MyD88) and Toll-IL-1-resistance (TIR) domain-containing adaptor-inducing IFN-beta (TRIF). Six hours after intraperitoneal injection of highly purified LPS (UP-LPS, 5 mg/kg), in vivo gastrointestinal transit and intestinal muscularis gene transcripts of inflammatory mediators chemokine (C-X-C motif) ligand 10, synonymous IP-10 (CXCL10), granulomonocyte colony stimulating factor (GM-CSF, synonymous CSF-2), IL-1beta, IL-6, IL-10, and inducible NO synthase (iNOS) were assessed in mice with transgenic loss-of-function for MyD88 or TRIF. LPS-induced MyD88 and TRIF mRNA upregulation was quantified within the intestinal muscularis of TLR4-competent and TLR4-mutant mice, and MyD88 mRNA levels were additionally measured in TLR4 bone marrow chimeras. MyD88 deficiency completely protected mice from early endotoxin-induced ileus, while TRIF deficiency partially ameliorated ileus severity. LPS induction of the primary downstream signaling element MyD88 was TLR4 dependent and was derived in equal amounts from both the hematopoietic and the nonhematopoietic cells. Conversely, no induction of TRIF mRNA was detectable. Significant gene induction of all inflammatory mediators was dependent on intracellular signal transduction by MyD88, while the TRIF MyD88-independent pathway predominantly regulated the molecular levels of CXCL10. In summary, MyD88 and TRIF are nonredundant signaling pathways in early endotoxin-induced rodent ileus, but MyD88 is the essential adaptor molecule for transduction of early TLR4-induced ileus and inflammatory signaling. The dependency of ileus on individual adaptor protein pathways is also reflected in the manifestation of specific molecular inflammatory events within the intestinal muscularis externa.
Collapse
Affiliation(s)
- Bettina M. Buchholz
- 1Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, Pennsylvania; ,2Department of Surgery, Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany; and
| | - Timothy R. Billiar
- 3Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anthony J. Bauer
- 1Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, Pennsylvania;
| |
Collapse
|
47
|
Peritoneal damage: the inflammatory response and clinical implications of the neuro-immuno-humoral axis. World J Surg 2010; 34:704-20. [PMID: 20049432 DOI: 10.1007/s00268-009-0382-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The peritoneum is a bilayer serous membrane that lines the abdominal cavity. We present a review of peritoneal structure and physiology, with a focus on the peritoneal inflammatory response to surgical injury and its clinical implications. METHODS We conducted a nonsystematic clinical review. A search of the Ovid MEDLINE database from 1950 through January 2009 was performed using the following search terms: peritoneum, adhesions, cytokine, inflammation, and surgery. RESULTS The peritoneum is a metabolically active organ, responding to insult through a complex array of immunologic and inflammatory cascades. This response increases with the duration and extent of injury and is central to the concept of surgical stress, manifesting via a combination of systemic effects, and local neural pathways via the neuro-immuno-humoral axis. There may be a decreased systemic inflammatory response after minimally invasive surgery; however, it is unclear whether this is due to a reduced local peritoneal reaction. CONCLUSIONS Interventions that dampen the peritoneal response and/or block the neuro-immuno-humoral pathway should be further investigated as possible avenues of enhancing recovery after surgery, and reducing postoperative complications.
Collapse
|
48
|
Moore BA, Peffer N, Pirone A, Bassiri A, Sague S, Palmer JM, Johnson DL, Nesspor T, Kliwinski C, Hornby PJ. GLP-2 receptor agonism ameliorates inflammation and gastrointestinal stasis in murine postoperative ileus. J Pharmacol Exp Ther 2010; 333:574-83. [PMID: 20167840 DOI: 10.1124/jpet.109.161497] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucagon-like peptide 2 (GLP-2) is a pleiotropic intestinotrophic hormone that we hypothesized could lessen gastrointestinal inflammation associated with postoperative ileus (POI). To test this idea, the prophylactic timing and dose of a long-acting variant of human GLP-2 linked to the Fc portion of murine immunoglobulin G (IgG) (GLP-2/IgG) was optimized in a murine model of POI. Surgically treated mice received a single dose of GLP-2/IgG, IgG isotype control, or phosphate-buffered saline 1 to 48 h before small bowel surgical manipulation. The distribution of orally fed fluorescein isothiocyanate-dextran and histological analyses of myeloperoxidase-positive immune cells were determined 24 and 48 h postoperatively. TaqMan quantitative polymerase chain reaction was used to determine early changes in mRNA expression in the muscularis or mucosa. In normal mice, prolonged exposure to GLP-2 increased upper gastrointestinal (GI) transit and mucosal weight. When administered 1 or 3 h before surgery, GLP-2/IgG reduced the leukocyte infiltrate 24 and 48 h postoperatively and improved GI transit 48 h postoperatively. Surgical manipulation rapidly increased gene expression of proinflammatory cytokines and enzymes for kinetically active mediators in the mucosa and muscularis. GLP-2/IgG2a affected the expression of genes associated with mucosal inflammation and barrier function. We conclude that prophylactic treatment with a long-acting GLP-2 agonist ameliorates inflammation and improves intestinal dysmotility associated with surgical manipulation of the bowel. The action of GLP-2 is consistent with a lessening of inflammation, leading to a more rapid recovery.
Collapse
Affiliation(s)
- Beverley A Moore
- Johnson & Johnson Pharmaceutical Research and Development, Spring House, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
De Backer O, Elinck E, Priem E, Leybaert L, Lefebvre RA. Peroxisome proliferator-activated receptor gamma activation alleviates postoperative ileus in mice by inhibition of Egr-1 expression and its downstream target genes. J Pharmacol Exp Ther 2009; 331:496-503. [PMID: 19657050 DOI: 10.1124/jpet.109.155135] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Postoperative ileus, a major cause of morbidity after abdominal surgery, is characterized by intestinal dysmotility and a complex inflammatory cascade within the intestinal muscularis. Treatment with carbon monoxide (CO)--inhaled or intraperitonea--has been shown to ameliorate bowel dysmotility caused by surgical manipulation of the gut in experimental animals. Recent evidence indicates that CO exerts its anti-inflammatory effects through the induction of peroxisome proliferator-activated receptor (PPAR)-gamma, a nuclear receptor whose activation has been linked to several physiological pathways, including those related to the regulation of intestinal inflammation. The purpose of this study was to evaluate pharmacological activation of PPARgamma in a murine model of postoperative ileus by use of the PPARgamma agonist rosiglitazone. Postoperative bowel dysmotility was induced by surgical manipulation of the colon. The functional severity of postoperative ileus was significantly ameliorated in mice pretreated with rosiglitazone (0.3 to 10 mg/kg i.p.); this was associated with a down-regulation of pro-inflammatory cytokines/chemokines, inducible nitric oxide synthase activity, cyclooxygenase-2 activity, as well as a decrease in leukocyte recruitment into the muscularis of both colon and jejunum. These anti-inflammatory effects were preceded by a PPARgamma-dependent down-regulation of early growth response (Egr)-1, a key regulator of inflammatory gene expression. In conclusion, these results indicate that rosiglitazone significantly attenuates postoperative ileus in mice by suppression of the muscularis inflammatory cascade through a PPARgamma-dependent down-regulation of Egr-1 and encourage the further clinical evaluation of synthetic PPARgamma agonists as pharmacological tool to prevent this postoperative event.
Collapse
Affiliation(s)
- Ole De Backer
- Heymans Institute of Pharmacology, Ghent University, B-9000 Ghent, Belgium.
| | | | | | | | | |
Collapse
|
50
|
Buchholz BM, Chanthaphavong RS, Bauer AJM. Nonhemopoietic cell TLR4 signaling is critical in causing early lipopolysaccharide-induced ileus. THE JOURNAL OF IMMUNOLOGY 2009; 183:6744-53. [PMID: 19846874 DOI: 10.4049/jimmunol.0901620] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Endotoxin-mediated ileus is poorly understood. Our objective was to mechanistically investigate the role of cell-specific TLR4 expression/signaling in causing gastrointestinal dysmotility. TLR4 chimeras and CSF-1-dependent macrophage-deficient mice were subjected to i.p. ultrapure (UP)-LPS (5 mg/kg). At 6 h, gastric emptying and gastrointestinal transit assessed in vivo motility, and jejunal circular muscle contractility was measured in vitro. Muscularis infiltration of neutrophils and monocytes were counted, and intestinal muscularis inflammatory mediators were quantified by quantitative PCR. Demonstrating TLR4 dependency, UP-LPS-induced gastric stasis and ileus of TLR4(WT) mice were absent in mutant TLR4(LPS-d) mice. Unexpectedly, engraftment of TLR4-mutant bone marrow into TLR4-competent mice (bmTLR4(LPS-d)/TLR4(WT)) exhibited a significant transit delay to UP-LPS similar to bmTLR4(WT)/TLR4(WT) mice. CSF-1(-/-) mice were not protected from ileus. Contrary, UP-LPS-treated bmTLR4(WT)/TLR4(LPS-d) and bmTLR4(LPS-d)/TLR4(LPS-d) mice had normal transit. No leukocytic infiltration was detected at 6 h. Spontaneous jejunal contractions were markedly suppressed in UP-LPS-treated TLR4-competent mice, but bethanechol-stimulated contractions were not altered by UP-LPS in any group. UP-LPS-induced inflammatory mRNAs in a TLR4-dependent manner, but TLR4 mRNA itself was not significantly altered. In chimera mice, UP-LPS induction of IL-1beta and IL-10 were hemopoietic dependent, and GM-CSF was nonhemopoietic dependent, whereas IL-6 and inducible NO synthase were derived from both cell types. Hemopoietic and nonhemopoietic cells contribute to TLR4-sensitive muscularis inflammatory signaling, but nonhemopoietic TLR4 signaling plays an exclusive primary role in causing functional UP-LPS-induced gastric stasis and ileus. Direct LPS suppression of spontaneous contractility participates in mediating early TLR4-transduced dysmotility.
Collapse
Affiliation(s)
- Bettina M Buchholz
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|