1
|
Hou M, Li X, Chen F, Tan Z, Han X, Liu J, Zhou J, Shi Y, Zhang J, Lv J, Leng Y. Naringenin alleviates intestinal ischemia/reperfusion injury by inhibiting ferroptosis via targeting YAP/STAT3 signaling axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156095. [PMID: 39383632 DOI: 10.1016/j.phymed.2024.156095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Intestinal ischemia/reperfusion injury (IRI) is a significant clinical emergency, and investigating novel therapeutic approaches and understanding their underlying mechanisms is essential for improving patient outcomes. Naringenin (Nar), a flavanone present in tomatoes and citrus fruits, is frequently consumed in the human diet and recognized for having immunomodulatory, anti-inflammatory, and antioxidant properties. Despite Nar being able to alleviate intestinal IRI, the exact molecular mechanisms remain elusive. PURPOSE To investigate Nar's protective properties on intestinal IRI and elucidate the mechanisms, a comprehensive approach that combines network pharmacology analysis with experimental verification in vitro and in vivo was adopted. METHODS The oxygen-glucose deprivation/reoxygenation (OGD/R) model in IEC-6 cells and a murine model of intestinal IRI were used. Nar's effects on intestinal IRI were assessed through histological analysis using H&E staining and tight junction (TJ) protein expression. Ferroptosis-related parameters, including iron levels, superoxide dismutase (SOD), glutathione (GSH), reactive oxygen species (ROS), malondialdehyde (MDA), and mitochondrial morphology, were analyzed. Network pharmacology was utilized to predict the pathways through which Nar exerts its anti-ferroptosis effects. Further mechanistic insights were obtained through si-RNA transfection, YAP inhibitor (verteporfin, VP) treatment, ferroptosis inhibitor (Ferrostatin-1) and ferroptosis inducer (Erastin) application, co-immunoprecipitation (Co-IP) and Western blotting. RESULTS Our results revealed that pretreatment with Nar significantly mitigated intestinal tissue damage and improved gut barrier function, as evidenced by increased TJ proteins (ZO-1 and Occludin). Nar reduced iron, MDA, and ROS, while it increased GSH and SOD levels. Additionally, Nar alleviated mitochondrial damage in mice. Nar treatment increased GPX4 and SLC7A11, while decreasing ACSL4 levels both in vivo and in vitro. Network pharmacology analysis suggested that Nar may target the Hippo signaling pathway. Notably, YAP, a key transcriptional co-activator within the Hippo pathway, was downregulated in intestinal IRI mice and OGD/R-induced IEC-6 cells. Nar pretreatment activated YAP, thereby augmenting anti-ferroptosis effects. The inhibition of YAP activation by VP or YAP knockdown increased p-STAT3 expression, thereby diminishing Nar's efficacy. Co-IP and immunofluorescence studies confirmed the interaction between YAP and STAT3. CONCLUSION This study shows that Nar can inhibit ferroptosis in intestinal IRI via activating YAP, which in turn suppresses STAT3 phosphorylation, thereby unveiling a novel mechanism and supporting Nar's potential to be a promising therapeutic agent for intestinal IRI.
Collapse
Affiliation(s)
- Min Hou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Xiaoxi Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Feng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Zhiguo Tan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Xiaoxia Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jie Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jia Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yajing Shi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jianmin Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jipeng Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China; Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
2
|
Akhigbe RE, Adedamola Aminat BO, Akhigbe TM, Hamed MA. Glutamine Alleviates I/R-Induced Intestinal Injury and Dysmotility Via the Downregulation of Xanthine Oxidase/Uric Acid Signaling and Lactate Generation in Wistar Rats. J Surg Res 2024; 295:431-441. [PMID: 38070257 DOI: 10.1016/j.jss.2023.11.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/14/2023] [Accepted: 11/13/2023] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Disruption of intestinal histoarchitecture and intestinal dysmotility is critical to intestinal ischemia/reperfusion (IR) injury and xanthine oxidase (XO)/uric acid (UA) signaling and increased lactate generation have been reported to play a role. More so, glutamine treatment has been demonstrated to inhibit XO/UA signaling. However, the role of glutamine in intestinal IR injury-induced intestinal dysmotility and the associated mechanisms of action are unclear. Therefore, this study was to investigate the mechanisms underlying the role of glutamine in intestinal IR injury. METHODS Forty male Wistar rats were acclimatized for two weeks and then randomized into four groups. The sham-operated, glutamine-treated, intestinal IR, and IR + glutamine groups. RESULTS Glutamine therapy attenuated the IR-induced increase in intestinal weight, disruption of intestinal histoarchitecture, and intestinal dysmotility. In addition, glutamine ameliorated IR-induced intestinal oxidative stress (increased malondialdehyde, reduced glutathione and superoxide dismutase, catalase, glutathione peroxidase, glutathione-S-transferase, and glucose-6-phosphate dehydrogenase activities), inflammation (increased TNF-α and IL-1β), and apoptosis (increased caspase three activity). These events were accompanied by glutamine alleviation of IR-induced upregulation of intestinal nuclear factor kappa B, XO/UA, and lactate generation. CONCLUSIONS In conclusion, XO/UA signaling and lactate levels are key factors in IR-induced intestinal injury and dysmotility, and glutamine-mediated XO/UA/lactate modulation may attenuate IR-induced intestinal injury and dysmotility.
Collapse
Affiliation(s)
- Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | | | - Tunmise Maryanne Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Breeding and Plant Genetics Unit, Department of Agronomy, Osun State University, Osun State
| | - Moses Agbomhere Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Medical Laboratory Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria; Department of Research and Bioinformatics, The Brainwill Laboratory, Osogbo, Osun State, Nigeria.
| |
Collapse
|
3
|
Zhao X, Liu D, Zhao Y, Wang Z, Wang Y, Chen Z, Ning S, Wang G, Meng L, Yao J, Tian X. HRD1-induced TMEM2 ubiquitination promotes ER stress-mediated apoptosis through a non-canonical pathway in intestinal ischemia/reperfusion. Cell Death Dis 2024; 15:154. [PMID: 38378757 PMCID: PMC10879504 DOI: 10.1038/s41419-024-06504-0] [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: 08/14/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a typical pathological course in the clinic with a high morbidity rate. Recent research has pointed out the critical role of ubiquitination during the occurrence and development of intestinal I/R by precisely mediating protein quality control and function. Here, we conducted an integrated multiomic analysis to identify critical ubiquitination-associated molecules in intestinal I/R and identified endoplasmic reticulum-located HRD1 as a candidate molecule. During intestinal I/R, excessive ER stress plays a central role by causing apoptotic pathway activation. In particular, we found that ER stress-mediated apoptosis was mitigated by HRD1 knockdown in intestinal I/R mice. Mechanistically, TMEM2 was identified as a new substrate of HRD1 in intestinal I/R by mass spectrometry analysis, which has a crucial role in attenuating apoptosis and promoting non-canonical ER stress resistance. A strong negative correlation was found between the protein levels of HRD1 and TMEM2 in human intestinal ischemia samples. Specifically, HRD1 interacted with the lysine 42 residue of TMEM2 and reduced its stabilization by K48-linked polyubiquitination. Furthermore, KEGG pathway analysis revealed that TMEM2 regulated ER stress-mediated apoptosis in association with the PI3k/Akt signaling pathway rather than canonical ER stress pathways. In summary, HRD1 regulates ER stress-mediated apoptosis through a non-canonical pathway by ubiquitinating TMEM2 and inhibiting PI3k/Akt activation during intestinal I/R. The current study shows that HRD1 is an intestinal I/R critical regulator and that targeting the HRD1/TMEM2 axis may be a promising therapeutic approach.
Collapse
Affiliation(s)
- Xuzi Zhao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Deshun Liu
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhao Chen
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Shili Ning
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Lu Meng
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China.
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China.
| |
Collapse
|
4
|
Wang X, Yan C, Wang C, Xu X, Liu Z, Wang X, Gong J. Protective effect of Cornuside on OGD/R injury in SH-SY5Y cells and its underlying mechanism. Brain Res 2023; 1821:148585. [PMID: 37722469 DOI: 10.1016/j.brainres.2023.148585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Apoptosis induced by oxygen-glucose deprivation/reperfusion (OGD/R) injury is the main cause of neuronal damage. Cornuside, a small-molecule cyclic enol ether terpene glycoside extracted from the dried fruit of mature Cornus officinalis Sieb. et Zucc., has vigorous anti-apoptotic and antioxidant effects. Previous studies have shown that Cornuside can reduce apoptosis and improve mitochondrial energy metabolism in cortical neurons of rats by inhibiting caspase-3 and calcium release. In this study, we treated SH-SY5Y cells with OGD/R to simulated ischemia/reperfusion (I/R) injury. Using high-throughput transcriptome sequencing, differentially expressed genes were analyzed in the OGD/R group versus the OGD/R + Cornuside (10 μmol/L) group to explore the neuroprotective mechanisms of Cornuside. The differentially expressed genes were mainly enriched in apoptosis signaling pathway, cell cycle, DNA damage and repair, and p38/JNK MAPK and p53 signaling pathways. The results showed that OGD/R significantly reduced the survival of SH-SY5Y cells, induced apoptosis, disrupted the nucleus, promoted the release of ROS, and led to cell cycle arrest. Cornuside reversed OGD/R-induced damage. By upregulating MAPK8IP1 and downregulating MAPK14, TP53INP1, and signaling pathway-related proteins (p-p38, p-JNK, and p-p53), Cornuside ameliorated cell damage induced by p38/JNK MAPK and p53 signaling pathways. Cornuside also downregulated apoptosis regulatory proteins (Bax, Bcl-2, caspase-3, caspase-9, and cytochrome c) and cell cycle regulatory proteins (cyclin B1, cyclin E, and p21).
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China; Department of Rehabilitation Medicine, Shengli Oilfield Central Hospital, Dongying 257097, PR China.
| | - Chao Yan
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China.
| | - Chaoyun Wang
- Department of Pharmacology, Binzhou Medical University, Yantai, 264003, PR China.
| | - Xiaoyan Xu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, PR China.
| | - Zhihao Liu
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China.
| | - Xin Wang
- Department of Rehabilitation Medicine, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| | - Jianwei Gong
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China.
| |
Collapse
|
5
|
De Pietro R, Martin J, Tradi F, Chopinet S, Barraud M, Gaudry M, Bourenne J, Nafati C, Boussen S, Guidon C, Bartoli M, Mege D. Prognostic factors after acute mesenteric ischemia: which patients require specific management? Int J Colorectal Dis 2023; 38:242. [PMID: 37777708 DOI: 10.1007/s00384-023-04540-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
PURPOSE Diagnosis and treatment of AMI are a real issue for implicating physicians. In the literature, only one AMI stroke center has reported its results so far, with increasing survival rates. Our aim was to analyze acute mesenteric ischemia (AMI) related mortality and predictive factors, in a single academic center, before creating a dedicated intestinal stroke center. METHODS All the patients with an AMI, between January 2015 and December 2020, were retrospectively included. They were divided into 2 groups according to the early mortality: death during the first 30 days and alive. The 2 groups were compared. RESULTS 173 patients (57% of men), were included, with a mean age of 68 ± 16 years. Overall mortality rate was 61%. Mortality occurred within the first 30 days in 78% of dead cases. Dead patients were significantly older, more frequently admitted from intensive care, with more serious clinical, laboratory and radiological characteristics. We have identified 3 protective factors - history of abdominal surgery (Odd Ratio = 0.1; 95%CI = 0.01-0.8, p = 0.03), medical management with curative anticoagulation (OR = 0.09; 95%CI = 0.02-0.5, p = 0.004) and/or antiplatelets (OR = 0.04; 95%CI = 0.006-0.3, p = 0.001)-, and 2 predictive factors of mortality - age > 70 years (OR = 7; 95%CI = 1.4-37, p = 0.02) and previous history of coronaropathy (OR = 13; 95%CI = 1.7-93, p = 0.01). CONCLUSIONS AMI is a severe disease with high morbidity and mortality rates. Even if its diagnosis is still difficult because of non-specific presentation, its therapeutic management needs to be changed in order to improve survival rates, particularly in patients older than 70 years with history of coronaropathy. Developing a dedicated organization would improve the diagnosis and the management of patients with AMI.
Collapse
Affiliation(s)
- Remi De Pietro
- Department of Digestive Surgery, Aix Marseille Univ, APHM, Timone University, 264 rue Saint-Pierre, 13005, Marseille, France
| | - Julie Martin
- Department of Emergency, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Farouk Tradi
- Department of Radiology, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Sophie Chopinet
- Department of Digestive Surgery, Aix Marseille Univ, APHM, Timone University, 264 rue Saint-Pierre, 13005, Marseille, France
| | - Marine Barraud
- Department of Gastro-enterology, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Marine Gaudry
- Department of Vascular Surgery, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Jeremy Bourenne
- Department of Emergency Critical Care Medicine, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Cyril Nafati
- Department of Intensive Care of Liver Diseases, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Salah Boussen
- Department of Intensive Care and Anesthesiology Department2, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Catherine Guidon
- Department of Anaesthesiology and Critical Care Medicine, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Michel Bartoli
- Department of Vascular Surgery, Aix Marseille Univ, APHM, Timone University, Marseille, France
| | - Diane Mege
- Department of Digestive Surgery, Aix Marseille Univ, APHM, Timone University, 264 rue Saint-Pierre, 13005, Marseille, France.
| |
Collapse
|
6
|
Tan C, Norden PR, Yu W, Liu T, Ujiie N, Lee SK, Yan X, Dyakiv Y, Aoto K, Ortega S, De Plaen IG, Sampath V, Kume T. Endothelial FOXC1 and FOXC2 promote intestinal regeneration after ischemia-reperfusion injury. EMBO Rep 2023; 24:e56030. [PMID: 37154714 PMCID: PMC10328078 DOI: 10.15252/embr.202256030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Pieter R Norden
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Wei Yu
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Naoto Ujiie
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Sun Kyong Lee
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Xiaocai Yan
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Kazushi Aoto
- Department of BiochemistryHamamatsu University School of MedicineHamamatsuJapan
| | - Sagrario Ortega
- Mouse Genome Editing Unit, Biotechnology ProgramSpanish National Cancer Research CentreMadridSpain
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Venkatesh Sampath
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
7
|
Cao Z, Mu S, Wang M, Zhang Y, Zou G, Yuan X, Huang Y, Yu S, Zhang J, Zhang C. Succinate pretreatment attenuates intestinal ischemia-reperfusion injury by inhibiting necroptosis and inflammation via upregulating Klf4. Int Immunopharmacol 2023; 120:110425. [PMID: 37285681 DOI: 10.1016/j.intimp.2023.110425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/21/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a common pathophysiological process in various diseases, and the disruption of the intestinal barrier composed of tight junction proteins is the initiating factor, which then leads to a large number of bacteria and endotoxins in the intestine into the bloodstream causing stress and distant organ damage. The release of inflammatory mediators and abnormal programmed death of intestinal epithelial cells are important factors of intestinal barrier damage. Succinate is an intermediate product of the tricarboxylic acid cycle with anti-inflammatory and pro-angiogenic activities, but its role in the maintenance of intestinal barrier homeostasis after I/R has not been fully elucidated. In this study, we explored the effect of succinate on intestinal ischemia-reperfusion injury and the possible mechanism of its role by flow cytometry, western blotting, real-time quantitative PCR and immunostaining. The results of pretreatment with succinate in the mouse intestinal I/R model and IEC-6 cells hypoxia-reoxygenation (H/R) model revealed a reduction in tissue damage, necroptosis and associated inflammation due to ischemia-reperfusion. Furthermore, it was found that the protective effect of succinate pretreatment may be associated with the transcriptional upregulation of the inflammatory protein KLF4 and the protective effect of intestinal barrier of succinate was diminished after inhibition of KLF4. Thus, our results suggest that succinate can exert a protective effect in intestinal ischemia-reperfusion injury through upregulation of KLF4 and also demonstrate the potential therapeutic value of succinate pretreatment in acute I/R injury of the intestine.
Collapse
Affiliation(s)
- Zhen Cao
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Silong Mu
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Maihuan Wang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yun Zhang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guijun Zou
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xinpu Yuan
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yun Huang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Siwang Yu
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
| | - Jinming Zhang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Chaojun Zhang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
8
|
Liu JF, Su G, Chen LX, Zhou JP, Gao J, Zhang JJ, Wu QH, Chen W, Chen DY, Zhang ZC. Irisin Attenuates Apoptosis Following Ischemia-Reperfusion Injury Through Improved Mitochondria Dynamics and ROS Suppression Mediated Through the PI3K/Akt/mTOR Axis. Mol Neurobiol 2023:10.1007/s12035-023-03336-5. [PMID: 37060502 DOI: 10.1007/s12035-023-03336-5] [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: 08/26/2022] [Accepted: 04/03/2023] [Indexed: 04/16/2023]
Abstract
Irisin is a muscle-derived hormone that promotes the survival of motor neurons and enhances muscle size following injury. In this study, we investigated the beneficial effects and mechanism(s) of action of irisin in response to cerebral ischemia-reperfusion injury (CIRI). Right-middle cerebral artery occlusion (MCAO) and hypoxia/reoxygenation (H/R) models were generated in C57BL/6 J mice. Mouse neuronal cell lines (NSC-34) were used to confirm the molecular mechanisms of the protection afforded by irisin in response to CIRI. We found that irisin (250 μg/kg) improved cerebral function and reduced the cerebral infarct volume following CIRI. Irisin also protected neuronal cells against ischemia-reperfusion (I/R) induced apoptosis, assessed via TUNEL, and cleaved Caspase-3 staining. Western blotting of neuronal tissue from irisin treated I/R mice showed lower expression of pro-apoptotic Bax and caspase-9 (P < 0.001 and P < 0.01) and increased levels of the pro-survival protein Bcl-2 (P < 0.01 & P < 0.001 vs. I/R). Irisin also reduced the levels of reactive oxygen species (ROS) characterized through malondialdehyde (MDA) assays. Irisin was found to maintain mitochondrial homeostasis through the suppression of mitochondrial fission-linked dynamin-related protein 1 in CIRI mice (P < 0.01 and P < 0.05 v. I/R cohort). Moreover, mitochondrial fusion-related protein (Mfn2) and Opa1 expression were rescued following irisin treatment (P < 0.001 and P < 0.01 v. I/R cohort). Cell-based assays showed that irisin activates PI3K/AKT/mTOR signaling in the neurons of CIRI mice. Furthermore, the beneficial effects of irisin on NSC-34 cell-survival, mitochondrial function, and ROS generation were reversed by VS-5584, a highly specific PI3K/AKT/mTOR inhibitor. Collectively, these data highlight the ability of irisin to alleviate CIRI in vivo and in vitro. The mechanisms of action of irisin include the attenuation of apoptosis through the prevention of mitochondrial fission and increased mitochondrial fusion and the alleviation of oxidative stress through activation of the PI3K/AKT/mTOR axis. We therefore identify irisin as a much-needed therapeutic for CIRI.
Collapse
Affiliation(s)
- Ji-Fei Liu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Li-Xia Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan-Ping Zhou
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Jia-Jia Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Qiong-Hui Wu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - De-Yi Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Zhen-Chang Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
9
|
Ala M, Fallahpour Khoshdel MR, Mohammad Jafari R, Sadrkhanloo M, Goudarzi S, Asl Soleimani M, Dehpour AR. Low-dose sumatriptan improves the outcome of acute mesenteric ischemia in rats via downregulating kynurenine. Pharmacol Rep 2023; 75:623-633. [PMID: 36920684 DOI: 10.1007/s43440-023-00470-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Mesenteric ischemia has remained without effective pharmacological management for many years. Sumatriptan, an abortive medication for migraine and cluster headaches, has potent anti-inflammatory properties and ameliorated organ ischemia in previous animal studies. Similarly, inhibition of the kynurenine pathway ameliorated renal and myocardial ischemia/reperfusion (I/R) in many preclinical studies. Herein, we assessed the effect of sumatriptan on experimental mesenteric I/R and investigated whether kynurenine pathway inhibition is a mechanism underlying its action. METHODS Ischemia was induced by ligating the origin of the superior mesenteric artery (SMA) and its anastomosis with the inferior mesenteric artery (IMA) with bulldog clamps for 30 min. Ischemia was followed by 1 h of reperfusion. Sumatriptan (0.1, 0.3, and 1 mg/kg ip) was injected 5 min before the reperfusion phase, 1-methyltryptophan (1-MT) (100 mg/kg iv) was used to inhibit kynurenine production. At the end of the reperfusion phase, samples were collected from the jejunum of rats for H&E staining and molecular assessments. RESULTS Sumatriptan improved the integrity of intestinal mucosa after I/R, and 0.1 mg/kg was the most effective dose of sumatriptan in this study. Sumatriptan decreased the increased levels of TNF-α, kynurenine, and p-ERK but did not change the decreased levels of NO. Furthermore, sumatriptan significantly increased the decreased ratio of Bcl2/Bax. Similarly, 1-MT significantly decreased TNF-α and kynurenine and protected against mucosal damage. CONCLUSIONS This study demonstrated that sumatriptan has protective effects against mesenteric ischemia and the kynurenine inhibition is potentially involved in this process. Therefore, it can be assumed that sumatriptan has the potential to be repurposed as a treatment for acute mesenteric ischemia.
Collapse
Affiliation(s)
- Moein Ala
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, 13145-784, 1416753955, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, 13145-784, Iran
| | - Mohammad Reza Fallahpour Khoshdel
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, 13145-784, 1416753955, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, 13145-784, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, 13145-784, 1416753955, Iran.
| | | | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, 13145-784, 1416753955, Iran
| | - Meisam Asl Soleimani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, 13145-784, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, 13145-784, 1416753955, Iran. .,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, 13145-784, Iran.
| |
Collapse
|
10
|
Gebran A, Dorken-Gallastegi A, Kaafarani HM. A Surgical Perspective of Gastrointestinal Manifestations and Complications of COVID-19 Infection. Gastroenterol Clin North Am 2023; 52:49-58. [PMID: 36813430 PMCID: PMC9537287 DOI: 10.1016/j.gtc.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2, has quickly spread over the world since December 2019. COVID-19 is a systemic disease that can affect various organs throughout the body. Gastrointestinal (GI) symptoms have been reported in 16% to 33% of all patients with COVID-19 and in 75% of critically ill patients. This chapter reviews the GI manifestations of COVID-19 as well as their diagnostic and treatment modalities.
Collapse
Affiliation(s)
| | | | - Haytham M.A. Kaafarani
- Corresponding author. Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, 165 Cambridge Street, Suite 810, Boston, MA 02114
| |
Collapse
|
11
|
Abstract
Acute mesenteric ischemia (AMI) is an uncommon yet highly lethal cause of acute abdomen in the emergency setting. Computed tomography (CT) imaging, in particular a biphasic protocol consisting of angiographic and venous phase scans, is widely used to corroborate non-specific clinical findings when suspicions of AMI are high. Techniques such as low kilovoltage peak scanning, dual energy acquisition, or a combined arterial/enteric phase can improve iodine conspicuity and evaluation of bowel enhancement. Biphasic CT with CT angiography is mandatory to directly assess for the 3 primary etiologies of AMI-arterial, venous, and non-occlusive mesenteric ischemia (NOMI), and the CT angiographic findings may be the first visible in the disease. In addition, numerous non-vascular CT findings have also been reported. Bowel wall thickening, mesenteric stranding, and ascites are common but non-specific findings that correlate poorly with disease severity. Pneumatosis intestinalis and portomesenteric venous gas, while not pathognomonic for ischemia, are highly specific in cases of high clinical suspicion. Bowel wall hypoenhancement is an early and specific sign but requires a protocol optimizing iodine conspicuity to confidently identify. Finally, intraperitoneal free air and solid organ infarcts are also highly specific ancillary findings in AMI. AMI occurs as a complication in 10% of small bowel obstruction (SBO) patients, and understanding imaging findings of ischemia in the context of SBO is necessary to aid in treatment planning and reduce over- and under-diagnosis of strangulation. Familiarity with the imaging features of ischemia by radiologists is vital to establish an early diagnosis before irreversible necrosis occurs.
Collapse
Affiliation(s)
- Hang Yu
- Department of Diagnostic Radiology, 8664University of Manitoba, Winnipeg, MB, Canada
| | - Iain D C Kirkpatrick
- Department of Diagnostic Radiology, 8664University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Aygun H, Olguner C, Koca U, Ergur BU, Sisman AR, Isguven D, Girgin P, Akkus M, Tulgar S. The effect of post-reperfusion levosimendan in an experimental intestinal ischemia-reperfusion model. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE (ONLINE) 2022; 2:45. [PMID: 37386547 DOI: 10.1186/s44158-022-00074-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/14/2022] [Indexed: 07/01/2023]
Abstract
BACKGROUND Levosimendan has been reported to have a positive effect on ischemia-reperfusion injury. Herein, we aimed to evaluate the effects of levosimendan applied after reperfusion in an experimental intestinal injury-reperfusion (IR) model. METHODS Twenty-one Wistar-albino male rats were separated into three groups: Sham group (n = 7): solely superior mesenteric artery (SMA) was dissected after laparotomy; intestinal ischemia-reperfusion group (IIR, n = 7): SMA was clamped for 60 min and unclamped for 120 min to cause ischemia-reperfusion; IIR + levosimendan group (IIR + L, n = 7): levosimendan was administered in ischemia-reperfusion model. The mean arterial pressures (MAP) were measured in all groups. MAP measurements were performed at the end of stabilization, at the 15th, 30th, and 60th minute of ischemia; at the 15th, 30th, 60th, and 120th minute of reperfusion; and at the end of levosimendan bolus application and when levosimendan infusion concluded. Reperfusion injury was evaluated with tissue malondialdehyde (MDA) and by Chiu score. RESULTS MAP at 15 min, 30 min, and 60 min of reperfusion was lower in IIR and IIR + L groups compared with basal inter-group measurements. Decline in MAP at 30 min after reperfusion was statistically significant in IIR and IIR + L groups when compared with the sham group. There was no significant difference between MDA levels in the groups. Chiu score was significantly lower in the sham group when compared to IIR and IIR + L groups and higher in IIR when compared to the IIR + L group. CONCLUSION Levosimendan leads to a decrease in intestinal damage although it did not affect lipid peroxidation and MAP when administered after reperfusion in an experimental intestinal IR model.
Collapse
Affiliation(s)
- Hakan Aygun
- Department of Anesthesiology, Bakircay University Cigli Training and Research Hospital, 8780/1 Sokak No:18 Yeni Mahalle Ata Sanayi, Izmir, Turkey.
| | - Cimen Olguner
- Department of Anesthesiology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ugur Koca
- Department of Anesthesiology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Bekir Ugur Ergur
- Department of Histology and Embryology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ali Rıza Sisman
- Department of Medical Biochemistry, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Duyguhan Isguven
- Department of Anesthesiology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Pelin Girgin
- Department of Anesthesiology, Bakircay University Cigli Training and Research Hospital, 8780/1 Sokak No:18 Yeni Mahalle Ata Sanayi, Izmir, Turkey
| | - Muhammed Akkus
- Department of Anesthesiology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Serkan Tulgar
- Department of Anesthesiology, Samsun University, Samsun Training and Research Hospital, Samsun, Turkey
| |
Collapse
|
13
|
Gebran A, El Moheb M, Argandykov D, Mashbari H, Gartland RM, Hwabejire JO, Velmahos GC, Kaafarani HM. Mesenteric Ischemia in Patients with Coronavirus 2019: A Scoping Review. Surg Infect (Larchmt) 2022; 23:781-786. [DOI: 10.1089/sur.2022.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Anthony Gebran
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mohamad El Moheb
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dias Argandykov
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hassan Mashbari
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Surgery, Faculty of Medicine, Jazan University, Saudi Arabia
| | - Rajshri M. Gartland
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John O. Hwabejire
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - George C. Velmahos
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
14
|
Wan Y, Dong P, Zhu X, Lei Y, Shen J, Liu W, Liu K, Zhang X. Bibliometric and visual analysis of intestinal ischemia reperfusion from 2004 to 2022. Front Med (Lausanne) 2022; 9:963104. [PMID: 36052333 PMCID: PMC9426633 DOI: 10.3389/fmed.2022.963104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a common tissue-organ damage occurring in surgical practice. This study aims to comprehensively review the collaboration and impact of countries, institutions, authors, subject areas, journals, keywords, and critical literature on intestinal I/R injury from a bibliometric perspective, and to assess the evolution of clustering of knowledge structures and identify hot trends and emerging topics. Methods Articles and reviews related to intestinal I/R were retrieved through subject search from Web of Science Core Collection. Bibliometric analyses were conducted on Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio). Results A total of 1069 articles and reviews were included from 2004 to 2022. The number of articles on intestinal I/R injury gradually plateaued, but the number of citations increased. These publications were mainly from 985 institutions in 46 countries, led by China and the United States. Liu Kx published the most papers, while Chiu Cj had the largest number of co-citations. Analysis of the journals with the most outputs showed that most journals focused on surgical sciences, cell biology, and immunology. Macroscopic sketch and microscopic characterization of the entire knowledge domain were achieved through co-citation analysis. The roles of cell death, exosomes, intestinal flora, and anesthetics in intestinal I/R injury are the current and developing research focuses. The keywords "dexmedetomidine", "proliferation", and "ferroptosis" may also become new trends and focus of future research. Conclusion This study comprehensively reviews the research on intestinal I/R injury using bibliometric and visualization methods, and will help scholars better understand the dynamic evolution of intestinal I/R injury and provide directions for future research.
Collapse
Affiliation(s)
- Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Xiaobing Zhu
- Department of Anesthesiology, Hospital of Traditional Chinese Medicine of Zhongshan City, Zhongshan, China
| | - Yuqiong Lei
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Shen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Weifeng Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Kexuan Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Xiyang Zhang
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Badripour A, Behzadi M, Hassanipour A, Azar PRS, Rahbar A, Abbaslou Z, Ehghaghi E, Piranviseh A, Khavandi MM, Ahmadi-Tafti SM, Ashouri M, Soltani ZE, Dehpour A. Albendazole ameliorates inflammatory response in a rat model of acute mesenteric ischemia reperfusion injury. Biomed Pharmacother 2022; 153:113320. [PMID: 35752010 DOI: 10.1016/j.biopha.2022.113320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute mesenteric ischemia is known as a life threatening condition. Re-establishment of blood flow in this condition can lead to mesenteric ischemia reperfusion (MIR) injury which is accompanied by inflammatory response. Still, clear blueprint of inflammatory mechanism underlying MIR injury has not been provided. Interestingly, Albendazole has exhibited notable effects on inflammation and cytokine production. In this study, we aimed to evaluate outcomes of MIR injury following pretreatment with Albendazole with respect to assessment of mesenteric inflammation and ischemia threshold. METHODS Male rats were randomly divided into sham operated, vehicle treated, Albendazole 100 mg/kg and Albendazole 200 mg/kg groups. MIR injury was induced by occlusion of superior mesenteric artery for 30 min followed by 120 min of reperfusion. Samples were utilized for assessment of epithelial survival and villous height. Immunohistochemistry study revealed intestinal expression of TNF-α and HIF-1-α. Gene expression of NF-κB/TLR4/TNF-α/IL-6 was measured using RTPCR. Also protein levels of inflammatory cytokines in serum and intestine were assessed by ELISA method. RESULTS Histopathological study demonstrated that pretreatment with Albendazole could ameliorate decline in villous height and epithelial survival following MIR injury. Also, systemic inflammation was suppressed after administration of Albendazole. Analysis of possible participating inflammatory pathway could demonstrate that intestinal expression of NF-κB/TLR4/TNF-α/IL-6 is significantly attenuated in treated groups. Eventually, IHC study illustrated concordant decline in mesenteric expression of HIF-1-α/TNF-α. CONCLUSION Single dose pretreatment with Albendazole could ameliorate inflammatory response and enhance ischemia threshold following induction of MIR injury. More studies would clarify existing causality in this phenomenon.
Collapse
Affiliation(s)
- Abolfazl Badripour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Behzadi
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Hassanipour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Pasha Reza Shams Azar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Rahbar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhaleh Abbaslou
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus Via Mersin 10, Turkey
| | - Elnaz Ehghaghi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus Via Mersin 10, Turkey
| | - Ashkan Piranviseh
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdi Khavandi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Ahmadi-Tafti
- Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran; Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ashouri
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahra Ebrahim Soltani
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Liao S, Luo J, Kadier T, Ding K, Chen R, Meng Q. Mitochondrial DNA Release Contributes to Intestinal Ischemia/Reperfusion Injury. Front Pharmacol 2022; 13:854994. [PMID: 35370747 PMCID: PMC8966724 DOI: 10.3389/fphar.2022.854994] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondria release many damage-associated molecular patterns (DAMPs) when cells are damaged or stressed, with mitochondrial DNA (mtDNA) being. MtDNA activates innate immune responses and induces inflammation through the TLR-9, NLRP3 inflammasome, and cGAS-STING signaling pathways. Released inflammatory factors cause damage to intestinal barrier function. Many bacteria and endotoxins migrate to the circulatory system and lymphatic system, leading to systemic inflammatory response syndrome (SIRS) and even damaging the function of multiple organs throughout the body. This process may ultimately lead to multiple organ dysfunction syndrome (MODS). Recent studies have shown that various factors, such as the release of mtDNA and the massive infiltration of inflammatory factors, can cause intestinal ischemia/reperfusion (I/R) injury. This destroys intestinal barrier function, induces an inflammatory storm, leads to SIRS, increases the vulnerability of organs, and develops into MODS. Mitophagy eliminates dysfunctional mitochondria to maintain cellular homeostasis. This review discusses mtDNA release during the pathogenesis of intestinal I/R and summarizes methods for the prevention or treatment of intestinal I/R. We also discuss the effects of inflammation and increased intestinal barrier permeability on drugs.
Collapse
Affiliation(s)
- Shishi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Luo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tulanisa Kadier
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Zhang JM, Wang KN, Zhang Y, Zhang JZ, Yuan XP, Zou GJ, Cao Z, Zhang CJ. BRCC36 promotes intestinal mucosal barrier injury caused by BMP2 after ischemia reperfusion via inhibiting PPARγ signaling. Biosci Biotechnol Biochem 2022; 86:331-339. [PMID: 34888627 DOI: 10.1093/bbb/zbab210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022]
Abstract
As one of the most common pathological changes in trauma and surgery practice, intestinal ischemia-reperfusion (I/R) injury is regarded as a major precipitating factor in the occurrence and development of fatal diseases. BRCA1-BRCA2-containing complex subunit 36 (BRCC36), a deubiquitinase, has been proved important in a variety of pathophysiological processes such as DNA repair, cell cycle regulation, tumorigenesis, and inflammatory response. However, the effect of BRCC36 on intestinal mucosal barrier injury after I/R has not been fully elucidated. Our research found that BRCC36 aggravated intestinal mucosal barrier injury caused by bone morphogenetic protein 2 after I/R by downregulating peroxisome proliferator-activated receptor-γ (PPARγ) signaling. These results suggested that BRCC36/PPARγ axis might serve as a potential therapeutic target for preventing intestinal mucosal barrier injury after I/R.
Collapse
Affiliation(s)
- Jin-Ming Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Kun-Nan Wang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Yun Zhang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jun-Ze Zhang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xin-Pu Yuan
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Gui-Jun Zou
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhen Cao
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chao-Jun Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
18
|
Srisajjakul S, Prapaisilp P, Bangchokdee S. Comprehensive review of acute small bowel ischemia: CT imaging findings, pearls, and pitfalls. Emerg Radiol 2022; 29:531-544. [PMID: 35122558 DOI: 10.1007/s10140-022-02028-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 01/12/2023]
Abstract
Acute small bowel ischemia is a life-threatening condition with a high mortality rate due to its lack of specific symptoms and laboratory profile, which render difficulty in establishing early diagnosis. The etiology of acute small bowel ischemia includes occlusive forms (arterial embolism, arterial thrombosis, and venous thrombosis) and nonocclusive mesenteric ischemia, of which arterial causes are far more common than venous causes. CT, the mainstay of accurate diagnoses, allows the identification of the features of vascular abnormalities and intestinal ischemic injuries, and helps clinicians to restore intestinal blood flow. Without treatment, the prognosis for acute small bowel ischemia is poor. A high index of suspicion and familiarity with the CT spectral findings of bowel ischemia are required to ensure rapid recognition of this condition.
Collapse
Affiliation(s)
- Sitthipong Srisajjakul
- Department of Radiology, Faculty of Medicine, Division of Diagnostic Radiology, Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Patcharin Prapaisilp
- Department of Radiology, Faculty of Medicine, Division of Diagnostic Radiology, Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Sirikan Bangchokdee
- Department of Internal Medicine, Pratumthani Hospital, 7 Ladlumkaew Muang District, Pratumthani, 12000, Thailand
| |
Collapse
|
19
|
Frontino G, Di Tonno R, Castorani V, Rigamonti A, Morotti E, Sandullo F, Scialabba F, Arrigoni F, Foglino R, Dionisi B, Ferri CIC, Zirpoli S, Barera G, Meschi F, Bonfanti R. Non-Occlusive Mesenteric Ischemia in Children With Diabetic Ketoacidosis: Case Report and Review of Literature. Front Endocrinol (Lausanne) 2022; 13:900325. [PMID: 35928892 PMCID: PMC9345499 DOI: 10.3389/fendo.2022.900325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Despite the use of technology, recurrent diabetic ketoacidosis (DKA) prevention remains an unmet need in children and adolescents with T1D and may be accompanied by life-threatening acute complications. We present a rare case of non-occlusive mesenteric ischemia (NOMI) with overt manifestation after DKA resolution and a discussion of recent literature addressing DKA-associated NOMI epidemiology and pathogenesis in children and adolescents. CASE PRESENTATION A 13-year-old female with previously diagnosed T1D, was admitted at our emergency department with hypovolemic shock, DKA, hyperosmolar state and acute kidney injury (AKI). Mildly progressive abdominal pain persisted after DKA correction and after repeated ultrasound evaluations ultimately suspect for intestinal perforation, an intraoperative diagnosis of NOMI was made. CONCLUSION The diagnosis of DKA-associated NOMI must be suspected in pediatric patients with DKA, persistent abdominal pain, and severe dehydration even after DKA resolution.
Collapse
Affiliation(s)
- Giulio Frontino
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Raffaella Di Tonno
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valeria Castorani
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
- *Correspondence: Valeria Castorani,
| | - Andrea Rigamonti
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Elisa Morotti
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federica Sandullo
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Scialabba
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesca Arrigoni
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Riccardo Foglino
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Benedetta Dionisi
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Salvatore Zirpoli
- Pediatric Radiology and Neuroradiology, Children’s Hospital “V. Buzzi”, Milan, Italy
| | - Graziano Barera
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Franco Meschi
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Riccardo Bonfanti
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
20
|
Hashmi SF, Rathore HA, Sattar MA, Johns EJ, Gan CY, Chia TY, Ahmad A. Hydrogen Sulphide Treatment Prevents Renal Ischemia-Reperfusion Injury by Inhibiting the Expression of ICAM-1 and NF-kB Concentration in Normotensive and Hypertensive Rats. Biomolecules 2021; 11:1549. [PMID: 34680182 PMCID: PMC8534271 DOI: 10.3390/biom11101549] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/13/2023] Open
Abstract
Our main objective was to investigate the effect of chronic administration of hydrogen sulphide donor (sodium hydrosulphide) on the expression of intercellular adhesion molecule-1 (ICAM-1) and concentration of nuclear factor-kappa B (NF-kB) in a renal ischemia-reperfusion injury (IRI) model of WKY and L-nitro-arginine-methyl-ester (L-NAME)-induced hypertensive rats. Sodium hydrosulphide (NaHS) was administered intraperitoneally (i.p.) for 35 days while cystathionine gamma lyase (CSE) inhibitor dL-propargylglycine (PAG) was administered at a single dose of 50 mg/kg. Animals were anesthetised using sodium pentobarbitone (60 mg/kg) and then prepared to induce renal ischemia by clamping the left renal artery for 30 min followed by 3 h of reperfusion. Pre-treatment with NaHS improved the renal functional parameters in both WKY and L-NAME-induced hypertensive rats along with reduction of blood pressure in hypertensive groups. Oxidative stress markers like malondialdehyde (MDA), total superoxide dismutase (T-SOD) and glutathione (GSH) were also improved by NaHS treatment following renal IRI. Levels of ICAM-1 and NF-kB concentration were reduced by chronic treatment with NaHS and increased by PAG administration after renal IRI in plasma and kidney. Treatment with NaHS improved tubular morphology and glomerulus hypertrophy. Pre-treatment with NaHS reduced the degree of renal IRI by potentiating its antioxidant and anti-inflammatory mechanism, as evidenced by decreased NF-kB concentration and downregulation of ICAM-1 expression.
Collapse
Affiliation(s)
- Syed F. Hashmi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Hassaan Anwer Rathore
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Munavvar A. Sattar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Edward J. Johns
- Department of Physiology, University College Cork, T12 K8AF Cork, Ireland;
| | - Chee-Yuen Gan
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Lebuh Bukit Jambul, Penang 11900, Malaysia;
| | - Tan Yong Chia
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Lebuh Bukit Jambul, Penang 11900, Malaysia;
| | - Ashfaq Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| |
Collapse
|
21
|
Filippone A, Casili G, Ardizzone A, Lanza M, Mannino D, Paterniti I, Esposito E, Campolo M. Inhibition of Prolyl Oligopeptidase Prevents Consequences of Reperfusion following Intestinal Ischemia. Biomedicines 2021; 9:biomedicines9101354. [PMID: 34680471 PMCID: PMC8533609 DOI: 10.3390/biomedicines9101354] [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] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Intestinal ischemia/reperfusion injury (IRI) remains a clinical event that contributes to high morbidity and mortality rates. Intestinal epithelium is exposed to histological and vascular changes following tissue ischemia. Prolyl endopeptidase (PREP), involved in inflammatory responses, could be targeted for recovery from the permanent consequences following intestinal ischemia. Our aim was to investigate the role of PREP inhibitor KYP-2047 in tissue damage, angiogenesis, and endothelial barrier permeability after intestinal IRI in mice. METHODS KYP-2047 treatments were performed 5 min prior to intestinal damage. Intestinal IRI was induced in mice by clamping the superior mesenteric artery and the celiac trunk for 30 min, followed by 1 h of reperfusion. RESULTS PREP inhibition by KYP-2047 treatment reduced intestinal IR-induced histological damage and neutrophil accumulation, limiting inflammation through decrease of NF-ĸB nuclear translocation and fibrotic processes. KYP-2047 treatment restored barrier permeability and structural alteration following intestinal IRI, attenuating neovascular processes compromised by ischemia/reperfusion. Additionally, loss of epithelial cells during intestinal ischemia occurring by apoptosis was limited by KYP-2047 treatment, which showed strong effects counteracting apoptosis and DNA damage. CONCLUSIONS These findings provide the first evidence that PREP inhibition through KYP-2047 inhibitor use could be a validate strategy for resolving alterations of intestinal epithelium the pathophysiology of intestinal disease.
Collapse
|
22
|
Gharishvandi F, Shafaroodi H, Mohammad Jafari R, Abdollahi A, Pasalar P, Dehpour AR. The protective effect of α7-nACh receptor and its interaction with 5-HT1B/1D receptors in acute intestinal ischemia-reperfusion injury in rats. Fundam Clin Pharmacol 2021; 36:100-113. [PMID: 34061415 DOI: 10.1111/fcp.12705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/08/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022]
Abstract
Over the past decades, great attention has been given to the nervous system modulating effects on the immune response in inflammation-associated injuries, such as acute intestinal ischemia-reperfusion (IR). Recently, we proved the anti-inflammatory and antioxidant effects of 5-hydroxytryptamine (5-HT)1B/1D receptors in intestinal IR injury in rats. Also, the alpha7 nicotinic acetylcholine (α7-nACh) receptor has anti-inflammatory effects in different inflammation-associated injuries. Starting from these premises, we aimed to examine the function of the α7-nACh receptors and the functional interactions between the anti-inflammatory and antioxidant effects of α7-nACh and 5-HT1B/1D receptors in acute intestinal IR injury. To confirm the expression and localization of α7-nACh receptors on the ileum nerves, an immunofluorescence-based method was applied. Then, intestinal IR injury was induced by 30-min occlusion of superior mesenteric artery and reperfusion for 2 h in rats. Acute systemic administration of α7-nACh receptor agonist PNU-282987 and antagonist methyllycaconitine, and 5-HT1B/1D receptors agonist (sumatriptan) and antagonist (GR127, 935) were used in the model of intestinal IR injury. Finally, biochemical and histological parameters were assessed. Α7-nACh receptors were expressed by 9% on the ileum nerves. Likewise, activation of the α7-nACh receptor showed anti-inflammatory and antioxidant effects in intestinal IR injury but not as well as 5-HT1B/1D receptors. Interestingly, 5-HT1B/1D receptors via attenuation of glutamate (Glu) release indirectly activated the α7-nACh receptor and its protective effects against inflammation and oxidative stress. The protective effect of the α7-nACh receptor on intestinal IR injury was activated indirectly through the 5-HT1B/1D receptors' modulatory impact on Glu release.
Collapse
Affiliation(s)
- Fatemeh Gharishvandi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Abdollahi
- Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Imam Hospital Complex, Tehran, Iran
| | - Parvin Pasalar
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Brain and Spinal Injury Repair Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Gutiérrez-Sánchez G, García-Alonso I, Gutiérrez Sáenz de Santa María J, Alonso-Varona A, Herrero de la Parte B. Antioxidant-Based Therapy Reduces Early-Stage Intestinal Ischemia-Reperfusion Injury in Rats. Antioxidants (Basel) 2021; 10:antiox10060853. [PMID: 34071753 PMCID: PMC8226848 DOI: 10.3390/antiox10060853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/02/2022] Open
Abstract
Intestinal ischemia-reperfusion injury (i-IRI) is a rare disorder with a high mortality rate, resulting from the loss of blood flow to an intestinal segment. Most of the damage is triggered by the restoration of flow and the arrival of cytokines and reactive oxygen species (ROS), among others. Inactivation of these molecules before tissue reperfusion could reduce intestinal damage. The aim of this work was to analyze the preventive effect of allopurinol and nitroindazole on intestinal mucosal damage after i-IRI. Wag/RijHsd rats were subjected to i-IRI by clamping the superior mesenteric artery (for 1 or 2 h) followed by a 30 min period of reperfusion. Histopathological intestinal damage (HID) was assessed by microscopic examination of histological sections obtained from injured intestine. HID was increased by almost 20% by doubling the ischemia time (from 1 to 2 h). Nitroindazole reduced HID in both the 1 and 2 h period of ischemia by approximately 30% and 60%, respectively (p < 0.001). Our preliminary results demonstrate that nitroindazole has a preventive/protective effect against tissue damage in the early stages of i-IRI. However, to better understand the molecular mechanisms underlying this phenomenon, further studies are needed.
Collapse
Affiliation(s)
- Gaizka Gutiérrez-Sánchez
- Department of Surgery and Radiology and Physical Medicine, University of The Basque Country, ES48940 Leioa, Biscay, Spain; (G.G.-S.); (I.G.-A.); (J.G.S.d.S.M.)
| | - Ignacio García-Alonso
- Department of Surgery and Radiology and Physical Medicine, University of The Basque Country, ES48940 Leioa, Biscay, Spain; (G.G.-S.); (I.G.-A.); (J.G.S.d.S.M.)
- Interventional Radiology Research Group, Biocruces Bizkaia Health Research Institute, ES48903 Barakaldo, Biscay, Spain
| | - Jorge Gutiérrez Sáenz de Santa María
- Department of Surgery and Radiology and Physical Medicine, University of The Basque Country, ES48940 Leioa, Biscay, Spain; (G.G.-S.); (I.G.-A.); (J.G.S.d.S.M.)
| | - Ana Alonso-Varona
- Department of Cell Biology and Histology, University of The Basque Country, ES48940 Leioa, Biscay, Spain;
| | - Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, University of The Basque Country, ES48940 Leioa, Biscay, Spain; (G.G.-S.); (I.G.-A.); (J.G.S.d.S.M.)
- Interventional Radiology Research Group, Biocruces Bizkaia Health Research Institute, ES48903 Barakaldo, Biscay, Spain
- Correspondence:
| |
Collapse
|
24
|
Silva FS, Canêdo VSR, Abreu BJ, Oliveira MF. Responses of matrix metalloproteinases to hyperbaric oxygen treatment: changing for good or ill? Connect Tissue Res 2021; 62:249-262. [PMID: 32900238 DOI: 10.1080/03008207.2020.1821675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Hyperbaric oxygen (HBO2) is currently emerging as a promising therapeutic option for diseases involving impaired tissue repair and remodeling. In this regard, HBO2 has been shown to modulate signaling pathways responsible for matrix metalloproteinases (MMPs) regulation, which makes the MMPs interesting targets for investigation. However, the understanding regarding how HBO2 treatment affects the expression and activity of the MMP family members in different tissues and diseases needs to be clarified. The precise roles of MMPs in the physiopathology of various tissue repair disorders also remain unclear. Because of potential off-target systemic effects of the HBO2 on MMPs, researchers and physicians should carefully consider whether their patients could be affected adversely by HBO2 exposure. Aims: This narrative review provides an overview of MMP biology (structure, function, and regulation) and summarizes available data showing how MMPs respond to HBO2 in different tissues and pathologies, also highlighting possible mechanisms.
Collapse
Affiliation(s)
- Flávio S Silva
- Department of Health Sciences, Federal Rural University of the Semi-Arid (UFERSA), Mossoró, Brazil
| | - Vítor S R Canêdo
- Department of Health Sciences, Federal Rural University of the Semi-Arid (UFERSA), Mossoró, Brazil
| | - Bento J Abreu
- Department of Morphology, Federal University of Rio Grande Do Norte (UFRN), Natal, Brazil
| | - Moacir F Oliveira
- Department of Animal Sciences, Federal Rural University of the Semi-Arid (UFERSA), Mossoró, Brazil
| |
Collapse
|
25
|
Zhang Y, Khalique A, Du X, Gao Z, Wu J, Zhang X, Zhang R, Sun Z, Liu Q, Xu Z, Midgley AC, Wang L, Yan X, Zhuang J, Kong D, Huang X. Biomimetic Design of Mitochondria-Targeted Hybrid Nanozymes as Superoxide Scavengers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006570. [PMID: 33480459 DOI: 10.1002/adma.202006570] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/27/2020] [Indexed: 06/12/2023]
Abstract
Development of enzyme mimics for the scavenging of excessive mitochondrial superoxide (O2 •- ) can serve as an effective strategy in the treatment of many diseases. Here, protein reconstruction technology and nanotechnology is taken advantage of to biomimetically create an artificial hybrid nanozyme. These nanozymes consist of ferritin-heavy-chain-based protein as the enzyme scaffold and a metal nanoparticle core as the enzyme active center. This artificial cascade nanozyme possesses superoxide dismutase- and catalase-like activities and also targets mitochondria by overcoming multiple biological barriers. Using cardiac ischemia-reperfusion animal models, the protective advantages of the hybrid nanozymes are demonstrated in vivo during mitochondrial oxidative injury and in the recovery of heart functionality following infarction via systemic delivery and localized release from adhesive hydrogels (i.e., cardiac patch), respectively. This study illustrates a de novo design strategy in the development of enzyme mimics and provides a promising therapeutic option for alleviating oxidative damage in regenerative medicine.
Collapse
Affiliation(s)
- Yue Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Anila Khalique
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xinchen Du
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zhanxia Gao
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jin Wu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ran Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zhiyuan Sun
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Lianyong Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xiyun Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
- CAS Engineering Laboratory for Nanozymes, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Zhuang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- School of Medicine, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
26
|
Sampaio de Holanda G, Dos Santos Valença S, Maran Carra A, Lopes Lichtenberger RC, Franco OB, Ribeiro BE, Bittencourt Rosas SL, Santana PT, Lima Castelo-Branco MT, Pereira de Souza HS, Schanaider A. Sulforaphane and Albumin Attenuate Experimental Intestinal Ischemia-Reperfusion Injury. J Surg Res 2021; 262:212-223. [PMID: 33610056 DOI: 10.1016/j.jss.2021.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/28/2020] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) injury constitutes a severe disorder, in great part resulting from oxidative stress. Because sulforaphane and albumin were shown to increase antioxidant defenses, we evaluated the therapeutic potential of these agents in an experimental model of I/R injury. METHODS Wistar rats were used to establish a model of intestinal I/R (35 min of ischemia, followed by 45 min of reperfusion) and were treated with albumin (5 mL/kg), sulforaphane (500 μg/kg), or saline intravenously before reperfusion. Animals that were not subjected to I/R served as the sham (laparotomy only) and control groups. Blood samples were analyzed for arterial gas, reactive oxygen species, and reactive nitrogen species using different molecular fluorescent probes. After euthanasia, ileal samples were collected for analysis, including histopathology, immunohistochemistry, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling assays, and lactic dehydrogenase measurement. RESULTS Oxygenation status and hemodynamic parameters were uniform during the experiment. The sulforaphane- or albumin-treated groups showed reduced concentrations of reactive oxygen species (P < 0.04), nitric oxide (P < 0.001), and peroxynitrite (P = 0.001), compared with I/R injury untreated animals. Treatment with sulforaphane or albumin resulted in the preservation of goblet cells (P < 0.03), reductions in histopathologic scores (P < 0.01), macrophage density (P < 0.01), iNOS expression (P < 0.004), NF-kappa B activation (P < 0.05), and apoptotic rates (P < 0.04) in the mucosa and a reduction in the concentration of lactic dehydrogenase (P < 0.04), more pronounced with sulforaphane. CONCLUSIONS Attenuation of intestinal I/R injury in this model probably reflects the antioxidative effects of systemic administration of both sulforaphane and albumin and reinforces their use in future translational research.
Collapse
Affiliation(s)
- Gustavo Sampaio de Holanda
- Departamento de Cirurgia, Centro de Cirurgia Experimental, Programa de Pós-Graduação em Ciências Cirúrgicas, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Amabile Maran Carra
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Olavo Borges Franco
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Beatriz Elias Ribeiro
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | | | - Patricia Teixeira Santana
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | | | | | - Alberto Schanaider
- Departamento de Cirurgia, Centro de Cirurgia Experimental, Programa de Pós-Graduação em Ciências Cirúrgicas, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Gendy A, Soubh A, Al-Mokaddem A, Kotb El-Sayed M. Dimethyl fumarate protects against intestinal ischemia/reperfusion lesion: Participation of Nrf2/HO-1, GSK-3β and Wnt/β-catenin pathway. Biomed Pharmacother 2021; 134:111130. [PMID: 33348309 DOI: 10.1016/j.biopha.2020.111130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Dimethyl fumarate (DMFU), a known Nrf2 activator, has proven its positive effect in different organs against ischemia/reperfusion (Is/Re) injury. Nevertheless, its possible impact to modulate intestinal Is/Re-induced injury has not been previously demonstrated before. Hence, this study aimed to investigate DMFU mechanistic maneuver against intestinal Is/Re. METHODS To accomplish this goal, Wistar rats were allocated into four groups; Sham-operated (SOP), intestinal Is/Re (1 h/6 h), and 14 days pre-treated DMFU (15 and 25 mg/kg/day, p.o). RESULTS The mechanistic maneuver divulged that DMFU safeguarded the intestine partly via amplifying the expression/content of Nrf2 along with enhancing its downstream, HO-1 expression/content. In addition, DMFU lessened GSK-3β expression/content accompanied by enriching β-catenin expression/content. The antioxidant action was affirmed by enhancing total antioxidant capacity, besides reducing MDA, iNOS, and its by-product, NOx. The DMFU action entailed anti-inflammatory character manifested by down-regulation of expression/content NF-κB with subsequent rebating the contents of TNF-α, IL-1β, and P-selectin, as well as MPO activity. Moreover, DMFU had anti-apoptotic nature demonstrated through enriching Bcl-2 level and diminishing that of caspase-3. CONCLUSION DMFU purveyed tenable novel protective mechanisms and mitigated events associated with intestinal Is/Re mischief either in the lower or the high dose partly by amending of oxidative stress and inflammation through the modulation of Nrf2/HO-1, GSK-3β, and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Abdallah Gendy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt.
| | - Ayman Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza, 12566, Egypt
| | - Asmaa Al-Mokaddem
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed Kotb El-Sayed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Helwan, 11790, Egypt
| |
Collapse
|
28
|
Liu X, Chen Y, You B, Peng Y, Chen Y, Yang Z, Zhang Y, Chen J. Molecular mechanism mediating enteric bacterial translocation after severe burn: the role of cystic fibrosis transmembrane conductance regulator. BURNS & TRAUMA 2021; 9:tkaa042. [PMID: 33501367 PMCID: PMC7809362 DOI: 10.1093/burnst/tkaa042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/28/2020] [Indexed: 11/23/2022]
Abstract
Background Gut ischemia and hypoxia post severe burn leads to breakdown of intestinal epithelial
barrier and enteric bacterial translocation (EBT), resulting in serious complications,
such as systemic inflammatory response syndrome, sepsis and multiple organ failure.
Cystic fibrosis transmembrane conductance regulator (CFTR) is known to be downregulated
by hypoxia and modulate junctional complexes, which are crucial structures maintaining
the intestinal barrier. This study aimed to investigate whether CFTR plays a role in
both regulating the intestinal barrier and mediating EBT post severe burn, as well as
the signaling pathways involved in these processes. Methods An in vitro Caco-2 cell model subjected to hypoxic injury and an
in vivo mouse model with a 30% total body surface area full-thickness
dermal burn were established. DF 508 mice (mice with F508del CFTR gene mutation) were
used as an in vivo model to further demonstrate the role of CFTR in maintaining normal
intestinal barrier function. QRT-PCR, western blot, ELISA, TER assay and
immunofluorescence staining were used to detect the expression and localization of CFTR
and tight junction proteins, as well as the function of tight junctions. Results Our data indicated that, in Caco-2 cells, the hypoxia condition significantly reduced
CFTR expression; activated extracellular signal-regulated kinase and nuclear factor-κB
signaling; elevated secretion of inflammatory factors (tumor necrosis factor-α,
interleukin-1β and interleukin-8); downregulated zonula occludens-1, occludin and
E-cadherin expression; decreased transepithelial electrical resistance values; and led
to a cellular mislocation of ZO-1. More importantly, knockdown of CFTR caused similar
alterations. The upregulation of inflammatory factors and downregulation of tight
junction proteins (ZO-1 and occludin) induced by knockdown of CFTR could be reversed by
specific extracellular signal-regulated kinase or nuclear factor-κB inhibition. In
support of the in vitro data, exuberant secretion of pro-inflammatory
mediators and EBT was observed in the intestine of severely burnt mice in
vivo. EBT occurred in DF508 mice (mice with the F508del CFTR gene mutation),
accompanied by augmented tumor necrosis factor-α, interleukin-1β and interleukin-8
levels in the ileum compared to wildtype mice. In addition, vitamin D3 was shown to
protect the intestinal epithelial barrier from hypoxic injury. Conclusions Collectively, the present study illustrated that CFTR and downstream signaling were
critical in modulating the intestinal epithelial junction and EBT post severe burn.
Collapse
Affiliation(s)
- Xinzhu Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China
| | - Yu Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China
| | - Bo You
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China.,Department of Burn and Plastic Surgery, No. 958 Hospital of Army, Southwest Hospital, Third Military Medical University (Army Military Medical University), Jian Xin Dong Street, Chongqing 400020, China
| | - Yuan Peng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Zhi Zao Ju Road, Shanghai 200011, China
| | - Yajie Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China
| | - Zichen Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Zhi Zao Ju Road, Shanghai 200011, China
| | - Jing Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Key Laboratory for Proteomics Disease, Institute of Burn Research, Southwest Hospital (the First Affiliated Hospital), Third Military Medical University (Army Military Medical University), Gao Tan Yan Street, Chongqing 400038, China
| |
Collapse
|
29
|
Cilostazol mitigates mesenteric ischemia/reperfusion-induced lung lesion: Contribution of PPAR-γ, NF-κB, and STAT3 crosstalk. Life Sci 2020; 266:118882. [PMID: 33310046 DOI: 10.1016/j.lfs.2020.118882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022]
Abstract
AIMS Cilostazol (Cilo), a phosphodiesterase-III inhibitor, has signified its efficacy against different ischemia/reperfusion (IS/RE) models. Nevertheless, it has not fully illuminated its potential effect against intestinal IS/RE-induced lung injury. Consequently, the study was fashioned to evaluate the feasible mechanism of action of Cilo against intestinal IS/RE-induced lung injury. MAIN METHODS Wistar rats were treated with Cilo (0.1 g/kg, p.o.) or with a vehicle for 14 days prior to IS/RE, induced by clamping of the superior mesenteric artery for 30 min with subsequent clamp removal for 2 h. KEY FINDINGS The mechanistic study disclosed that Cilo protected the two studied organs, viz., lung, and intestine partially by intensifying the expression/content of PPAR-γ accompanied by reducing the expression/content of NF-қB-p65 and STAT3. In addition to normalizing MDA, iNOS, and NOx, the Cilo antioxidant power was confirmed by intensifying tissues content of the total antioxidant capacity. With regard to the anti-inflammatory effect, Cilo reduced the effects of TNF-α, IL-6, and ICAM-1, which were reflected in MPO activity. Furthermore, Cilo had an anti-apoptotic attribute demonstrated by enhancing Bcl-2 content and lessening caspase-3 level. SIGNIFICANCE Cilo provided conceivable protective mechanisms to modulate events concomitant with mesenteric IS/RE partly by modulating oxidative stress, inflammation, and apoptosis feasibly via the participation of PPAR-γ, STAT3, and NF-κB p65 signaling pathways.
Collapse
|
30
|
Involvement of 5-HT1B/1D receptors in the inflammatory response and oxidative stress in intestinal ischemia/reperfusion in rats. Eur J Pharmacol 2020; 882:173265. [PMID: 32574671 DOI: 10.1016/j.ejphar.2020.173265] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
Acute mesenteric ischemia (AMI) is caused by an abrupt cessation of blood flow to the small intestine. Reperfusion is the return of blood flow to the ischemic bowel. Intestinal ischemia/reperfusion (I/R) leads to the formation of reactive oxygen species, local inflammatory response, and may lead to the patient's death. Pre-treatment of the intestinal may reduce the high mortality associated with AMI. 5-Hydroxytryptamine 1B (5-HT1B) and 5-HT1D receptors have anti-inflammatory and neuroprotective effects in different experimental studies. We aimed to investigate the potential involvement of these receptors in intestinal I/R injury. Firstly, we assessed the expression and localization of 5-HT1B and 5-HT1D receptors in the enteric nervous system using an immunofluorescence-based method. Intestinal I/R in rats was induced by 30 min occlusion of superior mesenteric artery and reperfusion for 2 h. Rats were randomly divided in different control and I/R groups (n = 6) receiving either vehicle, sumatriptan (5-HT1B/1D receptors agonist; 0.1 mg/kg), GR127,935 (5-HT1B/1D receptors antagonist; 0.1 mg/kg) and combination of sumatriptan (0.1 mg/kg) + GR127,935 (0.1 mg/kg) before determination of biochemical and histological parameters. In the enteric nervous system, 5-HT1B and 5-HT1D receptors were expressed 17% and 11.5%, respectively. Pre-treatment with sumatriptan decreased 5-hydroxytryptamine (5HT) level by 53%, and significantly decreased calcitonin gene-related peptide (CGRP) levels, lipid pereoxidation, neutrophil infiltration, and level of pro-inflammatory markers in the serum. Histopathologic studies also showed a remarkable decrease in intestinal tissue injury. These findings suggest that sumatriptan may inhibit intestinal injury induced by I/R through modulating the inflammatory response by activation of 5-HT1B/1D receptors.
Collapse
|
31
|
Schofield ZV, Wu MCL, Hansbro PM, Cooper MA, Woodruff TM. Acetate protects against intestinal ischemia‐reperfusion injury independent of its cognate free fatty acid 2 receptor. FASEB J 2020; 34:10418-10430. [DOI: 10.1096/fj.202000960r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Zoe V. Schofield
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
- The Institute for Molecular Bioscience The University of Queensland Brisbane QLD Australia
| | - Mike C. L. Wu
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
| | - Philip M. Hansbro
- Centre for Inflammation Centenary Institute Sydney NSW Australia
- Faculty of Science University of Technology Sydney Ultimo NSW Australia
| | - Matthew A. Cooper
- The Institute for Molecular Bioscience The University of Queensland Brisbane QLD Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
| |
Collapse
|
32
|
Amelioration of Coagulation Disorders and Inflammation by Hydrogen-Rich Solution Reduces Intestinal Ischemia/Reperfusion Injury in Rats through NF- κB/NLRP3 Pathway. Mediators Inflamm 2020; 2020:4359305. [PMID: 32587471 PMCID: PMC7303760 DOI: 10.1155/2020/4359305] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury often causes inflammatory responses and coagulation disorders, which is further promoting the deterioration of the disease. Hydrogen has anti-inflammatory, antioxidative, and antiapoptotic properties against various diseases. However, the effect of hydrogen on coagulation dysfunction after intestinal I/R and the underlying mechanism remains unclear. The purpose of this study was to explore whether hydrogen-rich solution (HRS) could attenuate coagulation disorders and inflammation to improve intestinal injury and poor survival following intestinal I/R. The rat model of intestinal I/R injury was established by clamping the superior mesenteric artery for 90 min and reperfusion for 2 h. HRS (10 or 20 mL/kg) or 20 mL/kg 0.9% normal saline was intravenously injected at 10 min before reperfusion, respectively. The samples were harvested at 2 h after reperfusion for further analyses. Moreover, the survival rate was observed for 24 h. The results showed that HRS improved the survival rate and alleviated serum diamine oxidase activities, intestinal injury, edema, and apoptosis. Interestingly, HRS markedly improved intestinal I/R-mediated coagulation disorders as evidenced by abnormal conventional indicators of coagulation and thromboelastography. Additionally, HRS attenuated inflammatory responses and the elevated tissue factor (TF) and inhibited nuclear factor kappa beta (NF-κB) and nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation in peripheral blood mononuclear cells. Moreover, inflammatory factors and myeloperoxidase were closely associated with TF level. This study thus emphasized upon the amelioration of coagulation disorders and inflammation by HRS as a mechanism to improve intestinal I/R-induced intestinal injury and poor survival, which might be partially related to inhibition of NF-κB/NLRP3 pathway.
Collapse
|
33
|
Soares ROS, Losada DM, Jordani MC, Évora P, Castro-E-Silva O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int J Mol Sci 2019; 20:ijms20205034. [PMID: 31614478 PMCID: PMC6834141 DOI: 10.3390/ijms20205034] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) permeates a variety of diseases and is a ubiquitous concern in every transplantation proceeding, from whole organs to modest grafts. Given its significance, efforts to evade the damaging effects of both ischemia and reperfusion are abundant in the literature and they consist of several strategies, such as applying pre-ischemic conditioning protocols, improving protection from preservation solutions, thus providing extended cold ischemia time and so on. In this review, we describe many of the latest pharmacological approaches that have been proven effective against IRI, while also revisiting well-established concepts and presenting recent pathophysiological findings in this ever-expanding field. A plethora of promising protocols has emerged in the last few years. They have been showing exciting results regarding protection against IRI by employing drugs that engage several strategies, such as modulating cell-surviving pathways, evading oxidative damage, physically protecting cell membrane integrity, and enhancing cell energetics.
Collapse
Affiliation(s)
| | - Daniele M Losada
- Department of Anatomic Pathology, Faculty of Medical Sciences, University of Campinas, 13083-970 Campinas, Brazil.
| | - Maria C Jordani
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Paulo Évora
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| | - Orlando Castro-E-Silva
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| |
Collapse
|
34
|
Toth S, Jonecova Z, Maretta M, Curgali K, Kalpakidis T, Pribula M, Kusnier M, Fagova Z, Fedotova J, La Rocca G, Rodrigo L, Caprnda M, Zulli A, Ciccocioppo R, Mechirova E, Kruzliak P. The effect of Betanin parenteral pretreatment on Jejunal and pulmonary tissue histological architecture and inflammatory response after Jejunal ischemia-reperfusion injury. Exp Mol Pathol 2019; 110:104292. [DOI: 10.1016/j.yexmp.2019.104292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/10/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
|
35
|
Wang C, Li Q, Ren J. Microbiota-Immune Interaction in the Pathogenesis of Gut-Derived Infection. Front Immunol 2019; 10:1873. [PMID: 31456801 PMCID: PMC6698791 DOI: 10.3389/fimmu.2019.01873] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived infection is among the most common complications in patients who underwent severe trauma, serious burn, major surgery, hemorrhagic shock or severe acute pancreatitis (SAP). It could cause sepsis and multiple organ dysfunction syndrome (MODS), which are regarded as a leading cause of mortality in these cases. Gut-derived infection is commonly caused by pathological translocation of intestinal bacteria or endotoxins, resulting from the dysfunction of the gut barrier. In the last decades, the studies regarding to the pathogenesis of gut-derived infection mainly focused on the breakdown of intestinal epithelial tight junction and increased permeability. Limited information is available on the roles of intestinal microbial barrier in the development of gut-derived infection. Recently, advances of next-generation DNA sequencing techniques and its utilization has revolutionized the gut microecology, leading to novel views into the composition of the intestinal microbiota and its connections with multiple diseases. Here, we reviewed the recent progress in the research field of intestinal barrier disruption and gut-derived infection, mainly through the perspectives of the dysbiosis of intestinal microbiota and its interaction with intestinal mucosal immune cells. This review presents novel insights into how the gut microbiota collaborates with mucosal immune cells to involve the development of pathological bacterial translocation. The data might have important implication to better understand the mechanism underlying pathological bacterial translocation, contributing us to develop new strategies for prevention and treatment of gut-derived sepsis.
Collapse
Affiliation(s)
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
36
|
Li S, Fujino M, Takahara T, Li XK. Protective role of heme oxygenase-1 in fatty liver ischemia-reperfusion injury. Med Mol Morphol 2019; 52:61-72. [PMID: 30171344 PMCID: PMC6542780 DOI: 10.1007/s00795-018-0205-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022]
Abstract
Ischemia-reperfusion (IR) injury is a kind of injury resulting from the restoration of the blood supply after blood vessel closure during liver transplantation and is the main cause of graft failure. The pathophysiological mechanisms of hepatic IR include a variety of oxidative stress responses. Hepatic IR is characterized by ischemia and hypoxia inducing oxidative stress, immune response and apoptosis. Fat-denatured livers are also used as donors due to the lack of liver donors. Fatty liver is less tolerant to IR than normal liver. Heme oxygenase (HO) is an enzyme that breaks down hemoglobin to bilirubin, ferrous iron and carbon monoxide (CO). Inducible HO subtype HO-1 is an important protective molecule in mammalian cells used to improve acute and chronic liver injury owing to its characteristic anti-inflammatory and anti-apoptotic qualities. HO-1 degrades heme, and its reaction product CO has been shown to reduce hepatic IR injury and increase the survival rate of grafts. As an induced form of HO, HO-1 also exerts a protective effect against liver IR injury and may be useful as a new strategy of ameliorating this kind of damage. This review summarizes the protective effects of HO-1 in liver IR injury, especially in fatty liver.
Collapse
Affiliation(s)
- Shaowei Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Terumi Takahara
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
37
|
Activation of G protein-coupled estrogen receptor protects intestine from ischemia/reperfusion injury in mice by protecting the crypt cell proliferation. Clin Sci (Lond) 2019; 133:449-464. [PMID: 30705108 DOI: 10.1042/cs20180919] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/14/2019] [Accepted: 01/30/2019] [Indexed: 12/13/2022]
Abstract
The intestinal ischemia/reperfusion (I/R) injury is a common clinical event related with high mortality in patients undergoing surgery or trauma. Estrogen exerts salutary effect on intestinal I/R injury, but the receptor type is not totally understood. We aimed to identify whether the G protein-coupled estrogen receptor (GPER) could protect the intestine against I/R injury and explored the mechanism. Adult male C57BL/6 mice were subjected to intestinal I/R injury by clamping (45 min) of the superior mesenteric artery followed by 4 h of intestinal reperfusion. Our results revealed that the selective GPER blocker abolished the protective effect of estrogen on intestinal I/R injury. Selective GPER agonist G-1 significantly alleviated I/R-induced intestinal mucosal damage, neutrophil infiltration, up-regulation of TNF-α and cyclooxygenase-2 (Cox-2) expression, and restored impaired intestinal barrier function. G-1 could ameliorate the impaired crypt cell proliferation ability induced by I/R and restore the decrease in villus height and crypt depth. The up-regulation of inducible nitric oxide synthase (iNOS) expression after I/R treatment was attenuated by G-1 administration. Moreover, selective iNOS inhibitor had a similar effect with G-1 on promoting the proliferation of crypt cells in the intestinal I/R model. Both GPER and iNOS were expressed in leucine-rich repeat containing G-protein coupled receptor 5 (Lgr5) positive stem cells in crypt. Together, these findings demonstrate that GPER activation can prompt epithelial cell repair following intestinal injury, which occurred at least in part by inhibiting the iNOS expression in intestinal stem cells (ISCs). GPER may be a novel therapeutic target for intestinal I/R injury.
Collapse
|
38
|
Ostjen CA, Rosa CGS, Hartmann RM, Schemitt EG, Colares JR, Marroni NP. Anti-inflammatory and antioxidant effect of melatonin on recovery from muscular trauma induced in rats. Exp Mol Pathol 2019; 106:52-59. [DOI: 10.1016/j.yexmp.2018.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/22/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
|
39
|
Yayla M, Cetin D, Adali Y, Kilicle PA, Toktay E. Potential therapeutic effect of pomegranate seed oil on ovarian ischemia/reperfusion injury in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 21:1262-1268. [PMID: 30627370 PMCID: PMC6312678 DOI: 10.22038/ijbms.2018.30149.7268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Objective(s): The aim of this study is to determine the therapeutic effects of pomegranate seed oil, which is a powerful antioxidant and anti-inflammatory agent, on ovarian-ischemia and reperfusion injury in rats. Materials and Methods: Fifty-six female albino Wistar rats were divided into 7 equal groups. Group 1; Sham Operation, Group 2; Ischemia, Group 3; Ischemia + Reperfusion, Group 4; Ischemia + Pomegranate 0,32 ml / kg (IP), Group 5; Ischemia + Pomegranate 0.64 ml / kg, Group 6; Ischemia + Pomegranate 0,32 ml / kg + reperfusion, Group 7; Ischemia + Pomegranate 0,64 ml / kg + reperfusion. Three hours after ischemia and 3 hours after reperfusion, the study was terminated. Results: While NADPH oxidase activity, MDA and TNF-α levels were significantly increased, SOD activity and GSH levels were reduced in ischemia and I/R groups. Low dose pomegranate seed oil application reduced significantly oxidative stress and NADPH oxidase activity in both ischemic and ischemic/reperfusion groups. At the same time, low-dose pomegranate seed oil extract reduced TNF-α levels and significantly increased antioxidant activity. Conclusion: PSO demonstrated an important therapeutic effect in the treatment of ovarian ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Muhammed Yayla
- Department of Pharmacology, Faculty of Medicine, Kafkas University, Kars, Turkey
| | - Damla Cetin
- Department of Pharmacology, Faculty of Medicine, Kafkas University, Kars, Turkey
| | - Yasemen Adali
- Department of Pathology, Highlited sentences should be cahanged as Canakkale Onsekiz Mart University Faculty of Medicine, 17100 Canakkale/Turkey
| | - Pinar Aksu Kilicle
- Department of Biology, Canakkale Onsekiz Mart University Faculty of Medicine, 17100 Canakkale/Turkey
| | - Erdem Toktay
- Department of Histology and Embryology, Ataturk University, Faculty of Medicine, 25240 Erzurum/Turkey
| |
Collapse
|
40
|
Zhang Z, Pu A, Yu M, Xiao W, Sun L, Cai Y, Yang H. Aryl hydrocarbon receptor activation modulates γδ intestinal intraepithelial lymphocytes and protects against ischemia/reperfusion injury in the murine small intestine. Mol Med Rep 2019; 19:1840-1848. [PMID: 30628695 DOI: 10.3892/mmr.2019.9823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/26/2018] [Indexed: 11/06/2022] Open
Abstract
The pathogenesis of intestinal ischemia/reperfusion (I/R) is associated with dysregulation of the intestinal immune system. The aryl hydrocarbon receptor (AhR), a receptor expressed in gamma‑delta (γδ) intraepithelial lymphocytes (IELs), is thought to regulate inflammation in the bowel. γδIELs are a key immunologic compartment with a capacity to modulate immune responses. In the present study, the function of the AhR in γδIELs in a mouse model of intestinal I/R injury was investigated to determine whether the AhR attenuates intestinal injury induced by intestinal I/R. Mice were assigned to three groups: sham, I/R and I/R+6‑formylindolo(3,2‑b)carbazole (FICZ). The sham group received no ischemia treatment, whereas the I/R and I/R+FICZ groups underwent upper mesenteric vessel ischemia for 30 min. The I/R group was injected intraperitoneally with 0.3 ml saline and the I/R+FICZ group was administered 1 µg of FICZ before a subsequent 6 h reperfusion. Then, the mice were sacrificed and the entire small intestinal tissues were collected for histologic examination. The phenotype and apoptosis of γδIELs and activation of CD4+ and CD8+ IELs were examined using flow cytometry. The cytokine mRNA and anti‑apoptosis gene expression in IELs were measured by qPCR. FICZ increased the γδIEL population and anti‑apoptosis genes in the γδIELs. FICZ reduced the percentage of activated CD4+ and CD8+ subpopulations and the expression of pro‑inflammatory mediator genes in IELs. FICZ inhibited inflammation in the gastrointestinal tract of mice with I/R injury. These results suggest that the AhR plays an important role in protecting the small intestine from I/R and increasing the γδIEL population by decreasing apoptosis of γδIELs.
Collapse
Affiliation(s)
- Zhicao Zhang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Aimin Pu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Yujiao Cai
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
41
|
Resveratrol Suppresses Gut-Derived NLRP3 Inflammasome Partly through Stabilizing Mast Cells in a Rat Model. Mediators Inflamm 2018; 2018:6158671. [PMID: 30670927 PMCID: PMC6317093 DOI: 10.1155/2018/6158671] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/07/2018] [Accepted: 11/04/2018] [Indexed: 12/13/2022] Open
Abstract
Background Inflammatory responses induced by intestinal ischemia-reperfusion (IIR) lead to serious systemic organ dysfunction and pose a challenge for current treatment. This study aimed at investigating the effects of resveratrol on IIR-induced intestinal injury and its influence on mast cells (MCs) in rats. Methods Rats subjected to intestinal ischemia for 60 min and 4 h of IIR were investigated. Animals were randomly divided into five groups (n = 8 per group): sham, IIR, resveratrol (RESV, 15 mg/kg/day for 5 days before operation) + IIR, cromolyn sodium (CS, MC membrane stabilizer) + IIR, and RESV + compound 48/80 (CP, MC agonist) + IIR. Results Intestinal injury and increased proinflammatory cytokines including tumor necrosis factor-α, interleukin-1β, and interleukin-18 were observed in the IIR group. Intestinal MC-related tryptase and β-hexosaminidase levels were also increased after rats were subjected to IIR accompanied by activation of NLRP3 inflammasomes. Interestingly, pretreatment with resveratrol significantly suppressed the activities of proinflammatory cytokines and attenuated intestinal injury. Resveratrol also reduced MC and NLRP3 inflammasome activation, which was consistent with the effects of cromolyn sodium. However, the protective effects of resveratrol were reversed by the MC agonist compound 48/80. Conclusions In summary, these findings reveal that resveratrol suppressed IIR injury by stabilizing MCs, preventing them from degranulation, accompanied with intestinal mucosa NLRP3 inflammasome inhibition and intestinal epithelial cell apoptosis reduction.
Collapse
|
42
|
Ren G, Yuan X, Zhao X, Hao Q, Cao J, Wang Y, Gao Q, Dou J, Zeng Q. Characterization and evolution of intestine injury at the anhepatic phase in portal hypertensive rats. Exp Ther Med 2018; 16:4765-4771. [PMID: 30542431 DOI: 10.3892/etm.2018.6800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 02/02/2018] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the characteristics and progression of intestinal injury at the anhepatic phase in portal hypertensive rats. A total of 120 healthy male Wistar rats were purchased, with 15 rats in the normal control group and 105 rats were assigned to establish a prehepatic portal hypertension model. The 105 model rats were further divided into seven treatment groups following ischemia-reperfusion. Meanwhile, portal vein pressure, the area of lower esophageal mucosal vein, endotoxin levels in portal vein blood and the level of malondialdehyde (MDA) and superoxide dismutase (SOD) were measured. Morphology changes of the intestine were observed using optical microscopy and transmission electron microscopy. A portal hypertension rat model was successfully established. Furthermore, endotoxin, MDA and SOD level reached a peak at 12-24 h following reperfusion and then decreased gradually to normal levels at 1 week following reperfusion. However, cytological damage did not recover to preoperative level within 1 week. These findings suggest that intestinal injury was most severe within 12-24 h following ischemia-reperfusion and most indicators recovered to almost normal levels. Therefore, further study on the intestinal mucosal damage is required, with the aim to reduce the production of intestinal endotoxin.
Collapse
Affiliation(s)
- Guijun Ren
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiaoye Yuan
- Department of Gerontology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Xin Zhao
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Qingchun Hao
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Jinglin Cao
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yang Wang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Qingjun Gao
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Jian Dou
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Qiang Zeng
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
43
|
Wang S, Xie T, Sun S, Wang K, Liu B, Wu X, Ding W. DNase-1 Treatment Exerts Protective Effects in a Rat Model of Intestinal Ischemia-Reperfusion Injury. Sci Rep 2018; 8:17788. [PMID: 30542063 PMCID: PMC6290768 DOI: 10.1038/s41598-018-36198-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
A growing number of studies have recently revealed a potential role for neutrophil extracellular traps (NETs) in the development of inflammation, coagulation and cell death. Deleterious consequences of NETs have been identified in ischemia-reperfusion (I/R)-induced organ damage, thrombosis and sepsis. And exogenous DNase-I has been suggested as a therapeutic strategy to attenuate ischemia-reperfusion (I/R) injuries in the kidney, brain and myocardium. Herein, we designed a study to investigate whether NETs contribute to the pathogenesis of intestinal I/R injury and evaluated the therapeutic value of DNase-1 in a rat model of intestinal I/R injury. In this rat model of intestinal I/R injury, we found that extracellular DNA was readily detectable in rat serum after 1 h of ischemia and 2 h of reperfusion. Treatment with DNase-1 significantly reduced the inflammatory response, restored intestinal barrier integrity and increased the expression of tight junction proteins. Our results indicate the existence of NETs in I/R-challenged intestinal tissues and firstly provide more evidence that DNase-1 may be an effective treatment for attenuating intestinal I/R injury.
Collapse
Affiliation(s)
- Shikai Wang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Tian Xie
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Shilong Sun
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Kai Wang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Baochen Liu
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Xingjiang Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China
| | - Weiwei Ding
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu Province, P.R. China.
| |
Collapse
|
44
|
Strand-Amundsen RJ, Tronstad C, Kalvøy H, Ruud TE, Høgetveit JO, Martinsen ØG, Tønnessen TI. Small intestinal ischemia and reperfusion-bioimpedance measurements. Physiol Meas 2018; 39:025001. [PMID: 29303488 DOI: 10.1088/1361-6579/aaa576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Trans-intestinal bioimpedance measurements have previously been used to investigate changes in electrical parameters during 6 h of ischemia in the small intestine. Knowledge is lacking regarding the time course of trans-intestinal bioimpedance parameters during reperfusion. As reperfusion is an important part in the clinical treatment of intestinal ischemia, we need to know how it affects the bioimpedance measurements. APPROACH We performed bioimpedance measurements, using a two-electrode setup on selected segments of the jejunum in 15 pigs. A controlled voltage signal was applied while measuring the resulting current. In each pig, five or six 30 cm segments of the jejunum were made ischemic by clamping the mesenteric arteries and veins creating segments with ischemia from 1-16 h duration. Reperfusion was initiated at selected time intervals of ischemia, and measured for 5-15 h afterwards. MAIN RESULTS The tan δ parameter (loss tangent) was different (p < 0.016) comparing ischemic and control tissue for the duration of the experiment (16 h). Comparing the control tissue 30 cm from the ischemic area with the control tissue 60 cm from the ischemic tissue, we found that the mean tan δ amplitude in the frequency range (3900-6300 Hz) was significantly higher (p < 0.036) in the proximal control after 10 h of experiment duration. After reperfusion, the time development of tanδm (loss tangent maximum over a frequency range) amplitude and frequency overlapped and periodically increased above the tanδm in the ischemic intestine. Dependent on the ischemic duration pre-reperfusion, the initial increase in tan δ stabilizes or increases drastically over time, compared to the tan δ amplitude of the ischemic tissue. SIGNIFICANCE As during ischemia, the electrical parameters during reperfusion also follow a characteristic time-course, depending on the ischemic exposure before pre-reperfusion. The temporal changes in electrical parameters during small intestinal ischemia followed by reperfusion provides important information for assessment of tissue injury.
Collapse
Affiliation(s)
- Runar J Strand-Amundsen
- Department of Clinical and Biomedical Engineering, Oslo University Hospital-Rikshospitalet, Postboks 4950 Nydalen, 0424 Oslo, Norway. Department of Physics, University of Oslo, Postboks 1048 Blindern, 0316 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
45
|
Ambrosi N, Guerrieri D, Caro F, Sanchez F, Haeublein G, Casadei D, Incardona C, Chuluyan E. Alpha Lipoic Acid: A Therapeutic Strategy that Tend to Limit the Action of Free Radicals in Transplantation. Int J Mol Sci 2018; 19:ijms19010102. [PMID: 29300330 PMCID: PMC5796052 DOI: 10.3390/ijms19010102] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/18/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022] Open
Abstract
Organ replacement is an option to mitigate irreversible organ damage. This procedure has achieved a considerable degree of acceptance. However, several factors significantly limit its effectiveness. Among them, the initial inflammatory graft reaction due to ischemia-reperfusion injury (IRI) has a fundamental influence on the short and long term organ function. The reactive oxygen species (ROS) produced during the IRI actively participates in these adverse events. Therapeutic strategies that tend to limit the action of free radicals could result in beneficial effects in transplantation outcome. Accordingly, the anti-oxidant α-lipoic acid (ALA) have been proved to be protective in several animal experimental models and humans. In a clinical trial, ALA was found to decrease hepatic IRI after hepatic occlusion and resection. Furthermore, the treatment of cadaveric donor and recipient with ALA had a protective effect in the short-term outcome in simultaneous kidney and pancreas transplanted patients. These studies support ALA as a drug to mitigate the damage caused by IRI and reinforce the knowledge about the deleterious consequences of ROS on graft injury in transplantation. The goal of this review is to overview the current knowledge about ROS in transplantation and the use of ALA to mitigate it.
Collapse
Affiliation(s)
- Nella Ambrosi
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
| | - Diego Guerrieri
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
| | - Fiorella Caro
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
| | - Francisco Sanchez
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
| | - Geraldine Haeublein
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
| | - Domingo Casadei
- Instituto de Nefrología de Buenos Aires, Nephrology, Buenos Aires C1199ABB, Argentina.
| | - Claudio Incardona
- GADOR SA, Buenos Aires C1199ABB, Argentina.
- Hospital Italiano, Buenos Aires C1199ABB, Argentina.
| | - Eduardo Chuluyan
- CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1199ABB, Argentina.
- Instituto de Nefrología de Buenos Aires, Nephrology, Buenos Aires C1199ABB, Argentina.
| |
Collapse
|
46
|
Verhaegh R, Petrat F, Brencher L, Kirsch M, de Groot H. Autodigestion by migrated trypsin is a major factor in small intestinal ischemia-reperfusion injury. J Surg Res 2017; 219:266-278. [DOI: 10.1016/j.jss.2017.05.084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/16/2017] [Accepted: 05/23/2017] [Indexed: 01/01/2023]
|
47
|
Turan I, Ozacmak HS, Ozacmak VH, Barut F, Araslı M. Agmatine attenuates intestinal ischemia and reperfusion injury by reducing oxidative stress and inflammatory reaction in rats. Life Sci 2017; 189:23-28. [PMID: 28893640 DOI: 10.1016/j.lfs.2017.08.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Inci Turan
- Department of Physiology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey.
| | - Hale Sayan Ozacmak
- Department of Physiology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - V Haktan Ozacmak
- Department of Physiology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Figen Barut
- Department of Pathology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Mehmet Araslı
- Department of Immunology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
48
|
Filiz S, Enis U, Mustafa I, Aysegul C, Elvan B. Protective Effect of Mesna on Intestinal Ischemia-reperfusion Injury by Nitric Oxide and Arginase in an Experimental Rat Model. INT J PHARMACOL 2017. [DOI: 10.3923/ijp.2017.1038.1046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
Pretreatment with the ALDH2 agonist Alda-1 reduces intestinal injury induced by ischaemia and reperfusion in mice. Clin Sci (Lond) 2017; 131:1123-1136. [PMID: 28325855 PMCID: PMC5434792 DOI: 10.1042/cs20170074] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/05/2017] [Accepted: 03/20/2017] [Indexed: 12/20/2022]
Abstract
Many studies demonstrate that activation of aldehyde dehydrogenase 2 (ALDH2) protects against oxidative stress via detoxification of cytotoxic aldehydes, and could attenuate cardiac, cerebral, lung and renal ischaemia-reperfusion (I/R) injuries. However, the effect of ALDH2 in intestinal I/R is unknown. The present study was set up to determine whether an ALDH2 agonist, Alda-1, could alleviate intestinal injury after gut I/R. In a mouse model of intestinal I/R injury, histological grading, proinflammatory cytokines, oxidative stress, cellular apoptosis, chemokine contents, ALDH2 activity, 4-hydroxy-trans-2-nonenal (4-HNE) and malondialdehyde (MDA) were evaluated. The results indicated that I/R treatment conferred elevation in pathological scores, proinflammatory cytokines, oxidative stress, cellular apoptosis and chemokine levels, accompanied by accumulated 4-HNE and MDA. No significant changes in ALDH2 activity were observed after I/R. However, Alda-1 pretreatment significantly decreased these injurious indicators, concomitant with up-regulated ALDH2 activity, and lessened 4-HNE and MDA accumulation. Taken together, our results implicate activation of ALDH2 by Alda-1 in the significant abatement intestinal I/R injury.
Collapse
|
50
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|