1
|
Xu W, Wang L, Chen R, Liu Y, Chen W. Pyroptosis and its role in intestinal ischemia-reperfusion injury: a potential therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04261-1. [PMID: 40372474 DOI: 10.1007/s00210-025-04261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Intestinal ischemia-reperfusion injury (II/RI) is a critical acute condition characterized by complex pathological mechanisms, including various modes of cell death. Among these, pyroptosis has garnered significant attention in recent years. This review explores the characteristics, molecular mechanisms, and implications of pyroptosis in II/RI, with a focus on therapeutic strategies targeting the pyroptosis pathway. Key processes such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation, caspase-1 activation, and gasdermin D (GSDMD)-mediated membrane pore formation are identified as central to pyroptosis. Compounds like MCC950, CY-09, metformin, and curcumin have shown promise in attenuating II/RI in preclinical studies by modulating these pathways. However, challenges remain in understanding non-canonical pyroptosis pathways, unraveling the exact mechanisms of GSDMD-induced pore formation, and translating these findings into clinical applications. Addressing these gaps will be crucial for developing innovative and effective treatments for II/RI.
Collapse
Affiliation(s)
- Wenping Xu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Lang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Ruili Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Yi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Wendong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
2
|
Canovai E, Farré R, De Hertogh G, Dubois A, Vanuytsel T, Pirenne J, Ceulemans LJ. Tranilast Reduces Intestinal Ischemia Reperfusion Injury in Rats Through the Upregulation of Heme-Oxygenase (HO)-1. J Clin Med 2025; 14:3254. [PMID: 40364289 PMCID: PMC12072342 DOI: 10.3390/jcm14093254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/15/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Intestinal ischemia reperfusion injury (IRI) is a harmful process that occurs during intestinal infarction and intestinal transplantation (ITx). It is characterized by severe inflammation which disrupts the mucosal barrier, causing bacterial translocation and sepsis. Tranilast (N-[3,4-dimethoxycinnamoyl]-anthranilic acid) (TL) is a synthetic compound with powerful anti-inflammatory properties. Objective: To investigate the effect of pretreatment with TL in a validated rat model of intestinal IRI (60 min of ischemia). Methods: TL (650 mg/kg) was administered by oral gavage 24 and 2 h before the onset of ischemia. Experiment 1 examined 7-day survival in 3 study groups (sham, vehicle+IRI and TL+IRI, n = 10/group). In Experiment 2, the effects on the intestinal wall integrity and inflammation were studied after 60 min of reperfusion using 3 groups (sham, IRI and TL+IRI, n = 6/group). The following end-points were studied: L-lactate, intestinal fatty acid-binding protein (I-FABP), histology, intestinal permeability, endotoxin translocation, pro- and anti-inflammatory cytokines and heme oxygenase-1 (HO-1) levels. Experiment 3 examined the role of HO-1 upregulation in TL pretreatment, by blocking its expression using Zinc protoporphyrin (ZnPP) at 20 mg/kg vs. placebo (n = 6/group). Results: Intestinal IRI resulted in severe damage of the intestinal wall and a 10% 7-day survival. These alterations led to endotoxin translocation and upregulation of pro-inflammatory cytokines. TL pretreatment improved survival up to 50%, significantly reduced inflammation and protected the intestinal barrier. The HO-1 inhibitor ZnPP, abolished the protective effect of TL. Conclusions: TL pretreatment improves survival by protecting the intestinal barrier function, decreasing inflammation and endotoxin translocation, through upregulation of HO-1.This rat study of severe intestinal ischemia reperfusion injury demonstrates a novel role for Tranilast as a potential therapy. Administration of Tranilast led to a marked reduction in mortality, inflammation and intestinal permeability and damage. The study proved that Tranilast functions through upregulation of heme oxygenase-1.
Collapse
Affiliation(s)
- Emilio Canovai
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Ricard Farré
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium;
| | - Gert De Hertogh
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Translational Cell and Tissue Research, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Antoine Dubois
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium;
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jacques Pirenne
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Laurens J. Ceulemans
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium; (E.C.); (G.D.H.); (A.D.); (T.V.); (J.P.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
3
|
Kaya C, Kapisiz A, Eryilmaz S, Karabulut R, Turkyilmaz Z, Inan MA, Aydin GY, Celik MA, Sonmez K. Effects of Lupeol on Intestinal Anastomosis After Experimental Intestinal Ischemia-Reperfusion Injury in Rats. Drug Des Devel Ther 2025; 19:479-490. [PMID: 39872634 PMCID: PMC11771168 DOI: 10.2147/dddt.s501289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/18/2025] [Indexed: 01/30/2025] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury can occur in a wide variety of diseases and surgeries. If necessary, the blood flow should be restored, including re-anastomosis by removing the intestines with impaired circulation. In this process, anastomotic strength is as important as inflammatory responses and oxidative stress. Therefore, we conducted the study to investigate the effects of lupeol on intestinal ischemia-reperfusion injury, not only biochemically and histopathologically but also on anastomotic strength and miRNAs. Methods Female rats were randomly divided into six groups. While only laparotomy was performed in the control group (Group C), anastomosis was performed in the sham group (Group S). In the other groups, the superior mesenteric artery was clamped for 45 minutes. In the groups I/R1 and L1, the intestine was transected, and end-to-end anastomosis was performed at the 1st hour of reperfusion. In the groups I/R24 and L24, this procedure was performed at the 24th hour of reperfusion. In addition, lupeol treatment was given before reperfusion and for the following 4 days in the groups L1 and L24. All rats, except the control group, bursting pressure was measured on the 5th day of anastomosis, and then all rats including the control group were sacrificed. TNF-α, IL-6 levels in blood samples and MDA, GSH, caspase-3, miR-29b-3p, miR-34a-5p, miR-495-3p levels in intestinal tissues were measured, and intestinal histopathology was also examined. Results Lupeol treatment, which was statistically significant in some parameters, demonstrated positive effects by decreasing TNF, IL-6, MDA, caspase-3, histopathological damage levels and increasing GSH and bursting pressure. In addition, lupeol decreased miR-34a-5p expression and increased miR-29b-3p and miR-495-3p expression. Conclusion Lupeol protected the intestines from I/R damage with its antioxidant and anti-inflammatory effects. Besides, it reduced the histopathological damage and increased the anastomotic strength. Additionally, miR-29b-3p, miR-34a-5p, miR-495-3p expressions were altered by lupeol.
Collapse
Affiliation(s)
- Cem Kaya
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Alparslan Kapisiz
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Sibel Eryilmaz
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Ramazan Karabulut
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Zafer Turkyilmaz
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Mehmet Arda Inan
- Department of Pathology, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | - Gizem Yaz Aydin
- Department of Biochemistry, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| | | | - Kaan Sonmez
- Department of Pediatric Surgery, Faculty of Medicine, Gazi University, Yenimahalle, Ankara, Turkey
| |
Collapse
|
4
|
Dubois A, Jin X, Hooft C, Canovai E, Boelhouwer C, Vanuytsel T, Vanaudenaerde B, Pirenne J, Ceulemans LJ. New insights in immunomodulation for intestinal transplantation. Hum Immunol 2024; 85:110827. [PMID: 38805779 DOI: 10.1016/j.humimm.2024.110827] [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: 02/16/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Tolerance is the Holy Grail of solid organ transplantation (SOT) and remains its primary challenge since its inception. In this topic, the seminal contributions of Thomas Starzl at Pittsburgh University outlined foundational principles of graft acceptance and tolerance, with chimerism emerging as a pivotal factor. Immunologically, intestinal transplantation (ITx) poses a unique hurdle due to the inherent characteristics and functions of the small bowel, resulting in increased immunogenicity. This necessitates heavy immunosuppression (IS) while IS drugs side effects cause significant morbidity. In addition, current IS therapies fall short of inducing clinical tolerance and their discontinuation has been proven unattainable in most cases. This underscores the unfulfilled need for immunological modulation to safely reduce IS-related burdens. To address this challenge, the Leuven Immunomodulatory Protocol (LIP), introduced in 2000, incorporates various pro-tolerogenic interventions in both the donor to the recipient, with the aim of facilitating graft acceptance and improving outcome. This review seeks to provide an overview of the current understanding of tolerance in ITx and outline recent advances in this domain.
Collapse
Affiliation(s)
- Antoine Dubois
- Unit of Abdominal Transplantation, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium; Abdominal Transplant Surgery, Department of Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Xin Jin
- Unit of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Charlotte Hooft
- Unit of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Emilio Canovai
- Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium; Oxford Transplant Centre, Churchill Hospital, Oxford, United Kingdom
| | - Caroline Boelhouwer
- Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium; Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (ChroMetA), KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Bart Vanaudenaerde
- Unit of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Unit of Abdominal Transplantation, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium; Abdominal Transplant Surgery, Department of Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT), University Hospitals Leuven, Leuven, Belgium; Unit of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Fleishman JS, Kumar S. Bile acid metabolism and signaling in health and disease: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:97. [PMID: 38664391 PMCID: PMC11045871 DOI: 10.1038/s41392-024-01811-6] [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: 11/28/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Collapse
Affiliation(s)
- Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
6
|
Khan H, Bangar A, Grewal AK, Singh TG. Mechanistic Implications of GSK and CREB Crosstalk in Ischemia Injury. Neurotox Res 2023; 42:1. [PMID: 38091155 DOI: 10.1007/s12640-023-00680-1] [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: 04/04/2023] [Revised: 11/03/2023] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
Ischemia-reperfusion (IR) injury is a damage to an organ when the blood supply is less than the demand required for normal functioning, leading to exacerbation of cellular dysfunction and death. IR injury occurs in different organs like the kidney, liver, heart, brain, etc., and may not only involve the ischemic organ but also cause systemic damage to distant organs. Oxygen-glucose deprivation in cells causes oxidative stress, calcium overloading, inflammation, and apoptosis. CREB is an essential integrator of the body's various physiological systems, and it is widely accepted that dysfunction of CREB signaling is involved in many diseases, including ischemia-reperfusion injury. The activation of CREB can provide life to a cell and increase the cell's survival after ischemia. Hence, GSK/CREB signaling pathway can provide significant protection to cells of different organs after ischemia and emerges as a futuristic strategy for managing ischemia-reperfusion injury. Different signaling pathways such as MAPK/ERK, TLR4/MyD88, RISK, Nrf2, and NF-κB, get altered during IR injury by the modulation of GSK-3 and CREB (cyclic AMP response element (CRE)-binding protein). GSK-3 (protein kinase B) and CREB are the downstream targets for fulfilling the roles of various signaling pathways. Calcium overloading during ischemia increases the expression of calcium-calmodulin-dependent protein kinase (CaMK), which subsequently activates CREB-mediated transcription, thus promoting the survival of cells. Furthermore, this review highlights the crosstalk between GSK-3 and CREB, promoting survival and rendering the cells resistant to subsequent severe ischemia.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Annu Bangar
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | | |
Collapse
|
7
|
Canovai E, Farré R, Accarie A, Lauriola M, De Hertogh G, Vanuytsel T, Pirenne J, Ceulemans LJ. INT-767-A Dual Farnesoid-X Receptor (FXR) and Takeda G Protein-Coupled Receptor-5 (TGR5) Agonist Improves Survival in Rats and Attenuates Intestinal Ischemia Reperfusion Injury. Int J Mol Sci 2023; 24:14881. [PMID: 37834329 PMCID: PMC10573246 DOI: 10.3390/ijms241914881] [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: 03/25/2023] [Revised: 09/25/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Intestinal ischemia is a potentially catastrophic emergency, with a high rate of morbidity and mortality. Currently, no specific pharmacological treatments are available. Previous work demonstrated that pre-treatment with obeticholic acid (OCA) protected against ischemia reperfusion injury (IRI). Recently, a more potent and water-soluble version has been synthesized: Intercept 767 (INT-767). The aim of this study was to investigate if intravenous treatment with INT-767 can improve outcomes after IRI. In a validated rat model of IRI (60 min ischemia + 60 min reperfusion), three groups were investigated (n = 6/group): (i) sham: surgery without ischemia; (ii) IRI + vehicle; and (iii) IRI + INT-767. The vehicle (0.9% NaCl) or INT-767 (10 mg/kg) were administered intravenously 15 min after start of ischemia. Endpoints were 7-day survival, serum injury markers (L-lactate and I-FABP), histology (Park-Chiu and villus length), permeability (transepithelial electrical resistance and endotoxin translocation), and cytokine expression. Untreated, IRI was uniformly lethal by provoking severe inflammation and structural damage, leading to translocation and sepsis. INT-767 treatment significantly improved survival by reducing inflammation and preserving intestinal structural integrity. This study demonstrates that treatment with INT-767 15 min after onset of intestinal ischemia significantly decreases IRI and improves survival. The ability to administer INT-767 intravenously greatly enhances its clinical potential.
Collapse
Affiliation(s)
- Emilio Canovai
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium (T.V.); (L.J.C.)
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Ricard Farré
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Alison Accarie
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Mara Lauriola
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
- Laboratory of Nephrology and Renal Transplantation, Department of Microbiology, Immunology, and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Gert De Hertogh
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium (T.V.); (L.J.C.)
- Translational Cell and Tissue Research, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium (T.V.); (L.J.C.)
- Translation Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
- Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jacques Pirenne
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium (T.V.); (L.J.C.)
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Laurens J. Ceulemans
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium (T.V.); (L.J.C.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
8
|
Elwany NE, Abdelhamid AM, Mohamed NM, Khalil SS, Elsayed Orabi EE, Abdelfattah AM. Vinpocetine alleviates intestinal ischemia/reperfusion injury and enhances M2 macrophage polarization in rats: Role of SIRT1/SOCS3/STAT3 signaling pathway. Int Immunopharmacol 2023; 122:110654. [PMID: 37459783 DOI: 10.1016/j.intimp.2023.110654] [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: 02/02/2023] [Revised: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 08/25/2023]
Abstract
Vinpocetine (Vinpo) is a neuroprotective vasodilator drug. It is an effective therapeutic agent for a variety of cerebrovascular and cognitive disorders. However, its potential protective efficacy on intestinal ischemia/reperfusion (I/R) injury remains elusive. The present study aimed to investigate the effect of Vinpo on intestinal I/R injury and to explore its modulatory effect on sirtuin (SIRT1)/ Suppressor of cytokine signaling (SOCS3)/ Signal Transducer and Activator of Transcription (STAT3) signaling. Twenty-four male Wistar albino rats were randomly allocated into four groups. G1 (sham): rats were subjected to surgical stress without I/R, GII (I/R): rats were subjected to 60 min/2-h I/R, GIII (Vinpo + I/R): rats were pre-treated with Vinpo (20 mg/kg/day, P.O. daily) for 2 weeks before intestinal I/R; GIV (EX527 + Vinpo + I/R): rats received both Vinpo (20 mg/kg/day, P.O.) and EX527 (5 mg/kg, once every 2 days, i.p) for 2 weeks before intestinal I/R. The current results showed that Vinpo improved the intestinal histopathological picture, enhanced M1 to M2 macrophage polarization and alleviated the I/R-induced increase in interleukins (IL-6, IL-1β), tumor necrosis factor (TNF-α), inducible nitric oxide synthase (i-NOS), and nitric oxide (NO). Additionally, Vinpo pretreatment upregulated SIRT1 mRNA expression/protein level and SOCS3 mRNA expression while downregulating P-STAT3 immunoreactivity. The effects of Vinpo were attenuated by the SIRT1 inhibitor EX527. We concluded that Vinpo ameliorated the intestinal I/R injury and enhanced M2 anti-inflammatory macrophage polarization through modulation of SIRT1/SOCS3/STAT3/i-NOS cascade.
Collapse
Affiliation(s)
- Nisreen E Elwany
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Egypt
| | | | | | - Sama S Khalil
- Medical Physiology Department, Faculty of Medicine, Zagazig University, Egypt
| | | | | |
Collapse
|
9
|
Clarysse M, Accarie A, Panisello-Roselló A, Farré R, Canovai E, Monbaliu D, De Hertogh G, Vanuytsel T, Pirenne J, Ceulemans LJ. Intravenous Polyethylene Glycol Alleviates Intestinal Ischemia-Reperfusion Injury in a Rodent Model. Int J Mol Sci 2023; 24:10775. [PMID: 37445954 DOI: 10.3390/ijms241310775] [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: 06/07/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Intestinal ischemia-reperfusion injury (IRI) is a common clinical entity, and its outcome is unpredictable due to the triad of inflammation, increased permeability and bacterial translocation. Polyethylene glycol (PEG) is a polyether compound that is extensively used in pharmacology as an excipient in various products. More recently, this class of products have shown to have potent anti-inflammatory, anti-apoptotic, immunosuppressive and cell-membrane-stabilizing properties. However, its effects on the outcome after intestinal IRI have not yet been investigated. We hypothesized that PEG administration would reduce the effects of intestinal IRI in rodents. In a previously described rat model of severe IRI (45 min of ischemia followed by 60 min of reperfusion), we evaluated the effect of IV PEG administration at different doses (50 and 100 mg/kg) before and after the onset of ischemia. In comparison to control animals, PEG administration stabilized the endothelial glycocalyx, leading to reduced reperfusion edema, bacterial translocation and inflammatory reaction as well as improved 7-day survival. These effects were seen both in a pretreatment and in a treatment setting. The fact that this product is readily available and safe should encourage further clinical investigations in settings of intestinal IRI, organ preservation and transplantation.
Collapse
Affiliation(s)
- Mathias Clarysse
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Alison Accarie
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Arnau Panisello-Roselló
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Cientificas (CSIC)-Institut D'Investigacions Biomèdique August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ricard Farré
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Emilio Canovai
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Diethard Monbaliu
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Gert De Hertogh
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
- Laboratory of Translational Cell & Tissue Research, KU Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jacques Pirenne
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
10
|
Yang JZ, Zhang KK, He JT, Chen LJ, Ding JF, Liu JL, Li JH, Liu Y, Li XW, Zhao D, Xie XL, Wang Q. Obeticholic acid protects against methamphetamine-induced anxiety-like behavior by ameliorating microbiota-mediated intestinal barrier impairment. Toxicology 2023; 486:153447. [PMID: 36720452 DOI: 10.1016/j.tox.2023.153447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/14/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
Methamphetamine (Meth) abuse can cause severe anxiety disorder and interfere with gut homeostasis. Obeticholic acid (OCA) has emerged as a protective agent against diet-related anxiety that improves gut homeostasis. The potential for OCA to ameliorate Meth-induced anxiety, and the microbial mechanisms involved, remain obscure. Here, C57/BL6 mice were intraperitoneally injected with Meth (15 mg/kg) to induce anxiety-like behavior. 16 S rRNA sequence analysis and fecal microbiome transplantation (FMT) were used to profile the gut microbiome and evaluate its effects, respectively. Orally administered OCA was investigated for protection against Meth-induced anxiety. Results indicated that Meth mediated anxiety-like behavior, aroused hippocampal neuroinflammation through activation of the TLR4/MyD88/NF-κB pathway, weakened intestinal barrier and disturbed the gut microbiome. Specifically, abundance of anxiety-related Rikenella was increased. FMT from Meth-administrated mice also weakened intestinal barrier and elevated serum LPS, inducing hippocampal neuroinflammation and anxiety-like behavior in recipient mice. Finally, OCA pretreatment ameliorated Meth-induced impairment of gut homeostasis by reshaping the microbial composition and improving the intestinal barrier. Meth-induced anxiety-like behavior and hippocampal neuroinflammation were also ameliorated by OCA pretreatment. These preliminary findings reveal the crucial role of gut microbiota in Meth-induced anxiety-like behavior and neuroinflammation, highlighting OCA as a potential candidate for the prevention of Meth-induced anxiety.
Collapse
Affiliation(s)
- Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kai-Kai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jie-Tao He
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, China
| | - Li-Jian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jin-Feng Ding
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jia-Hao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiu-Wen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Dong Zhao
- Key Laboratory of Evidence Science (China University of Political Science and Law), Ministry of Education, Beijing, China.
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong 510515, China.
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
11
|
Yang J, van Dijk TH, Koehorst M, Havinga R, de Boer JF, Kuipers F, van Zutphen T. Intestinal Farnesoid X Receptor Modulates Duodenal Surface Area but Does Not Control Glucose Absorption in Mice. Int J Mol Sci 2023; 24:ijms24044132. [PMID: 36835544 PMCID: PMC9961586 DOI: 10.3390/ijms24044132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/18/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
Bile acids facilitate the intestinal absorption of dietary lipids and act as signalling molecules in the maintenance of metabolic homeostasis. Farnesoid X receptor (FXR) is a bile acid-responsive nuclear receptor involved in bile acid metabolism, as well as lipid and glucose homeostasis. Several studies have suggested a role of FXR in the control of genes regulating intestinal glucose handling. We applied a novel dual-label glucose kinetic approach in intestine-specific FXR-/- mice (iFXR-KO) to directly assess the role of intestinal FXR in glucose absorption. Although iFXR-KO mice showed decreased duodenal expression of hexokinase 1 (Hk1) under obesogenic conditions, the assessment of glucose fluxes in these mice did not show a role for intestinal FXR in glucose absorption. FXR activation with the specific agonist GS3972 induced Hk1, yet the glucose absorption rate remained unaffected. FXR activation increased the duodenal villus length in mice treated with GS3972, while stem cell proliferation remained unaffected. Accordingly, iFXR-KO mice on either chow, short or long-term HFD feeding displayed a shorter villus length in the duodenum compared to wild-type mice. These findings indicate that delayed glucose absorption reported in whole-body FXR-/- mice is not due to the absence of intestinal FXR. Yet, intestinal FXR does have a role in the small intestinal surface area.
Collapse
Affiliation(s)
- Jiufang Yang
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Theo H. van Dijk
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
- Correspondence: (F.K.); (T.v.Z.); Tel.: +31-58-288-2132 (F.K.)
| | - Tim van Zutphen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The Netherlands
- Faculty Campus Fryslân, University of Groningen, 8911CE Leeuwarden, The Netherlands
- Correspondence: (F.K.); (T.v.Z.); Tel.: +31-58-288-2132 (F.K.)
| |
Collapse
|
12
|
Nevens F, Trauner M, Manns MP. Primary biliary cholangitis as a roadmap for the development of novel treatments for cholestatic liver diseases †. J Hepatol 2023; 78:430-441. [PMID: 36272496 DOI: 10.1016/j.jhep.2022.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022]
Abstract
The discovery of nuclear receptors and transporters has contributed to the development of new drugs for the treatment of cholestatic liver diseases. Particular progress has been made in the development of second-line therapies for PBC. These new drugs can be separated into compounds primarily targeting cholestasis, molecules targeting fibrogenesis and molecules with immune-mediated action. Finally, drugs aimed at symptom relief (pruritus and fatigue) are also under investigation. Obeticholic acid is currently the only approved second-line therapy for PBC. Drugs in the late phase of clinical development include peroxisome proliferator-activated receptor agonists, norursodeoxycholic acid and NADPH oxidase 1/4 inhibitors.
Collapse
Affiliation(s)
- Frederik Nevens
- Department of Gastroenterology and Hepatology, University Hospital KU Leuven, Belgium; Centre of ERN RARE-LIVER.
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Austria; Centre of ERN RARE-LIVER
| | - Michael P Manns
- Hannover Medical School, Hannover, Germany; Centre of ERN RARE-LIVER
| |
Collapse
|
13
|
Protective Effect of Oxygen and Isoflurane in Rodent Model of Intestinal Ischemia-Reperfusion Injury. Int J Mol Sci 2023; 24:ijms24032587. [PMID: 36768910 PMCID: PMC9917127 DOI: 10.3390/ijms24032587] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/22/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Animal research in intestinal ischemia-reperfusion injury (IRI) is mainly performed in rodent models. Previously, intraperitoneal (I.P.) injections with ketamine-xylazine mixtures were used. Nowadays, volatile anesthetics (isoflurane) are more common. However, the impact of the anesthetic method on intestinal IRI has not been investigated. We aim to analyze the different anesthetic methods and their influence on the extent of intestinal IRI in a rat model. Male Sprague-Dawley rats were used to investigate the effect of I.P. anesthesia on 60 min of intestinal ischemia and 60 min of reperfusion in comparison to hyperoxygenation (100% O2) and volatile isoflurane anesthesia. In comparison to I.P. anesthesia with room air (21% O2), supplying 100% O2 improved 7-day survival by cardiovascular stabilization, reducing lactic acidosis and preventing vascular leakage. However, this had no effect on the intestinal epithelial damage, permeability, and inflammatory response observed after intestinal IRI. In contrast to I.P. + 100% O2, isoflurane anesthesia reduced intestinal IRI by preventing ongoing low-flow reperfusion hypotension, limiting intestinal epithelial damage and permeability, and by having anti-inflammatory effects. When translating the aforementioned results of this study to clinical situations, such as intestinal ischemia or transplantation, the potential protective effects of hyperoxygenation and volatile anesthetics require further research.
Collapse
|
14
|
Gee LMV, Barron-Millar B, Leslie J, Richardson C, Zaki MYW, Luli S, Burgoyne RA, Cameron RIT, Smith GR, Brain JG, Innes B, Jopson L, Dyson JK, McKay KRC, Pechlivanis A, Holmes E, Berlinguer-Palmini R, Victorelli S, Mells GF, Sandford RN, Palmer J, Kirby JA, Kiourtis C, Mokochinski J, Hall Z, Bird TG, Borthwick LA, Morris CM, Hanson PS, Jurk D, Stoll EA, LeBeau FEN, Jones DEJ, Oakley F. Anti-Cholestatic Therapy with Obeticholic Acid Improves Short-Term Memory in Bile Duct-Ligated Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:11-26. [PMID: 36243043 DOI: 10.1016/j.ajpath.2022.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/03/2022] [Accepted: 09/28/2022] [Indexed: 12/12/2022]
Abstract
Patients with cholestatic liver disease, including those with primary biliary cholangitis, can experience symptoms of impaired cognition or brain fog. This phenomenon remains unexplained and is currently untreatable. Bile duct ligation (BDL) is an established rodent model of cholestasis. In addition to liver changes, BDL animals develop cognitive symptoms early in the disease process (before development of cirrhosis and/or liver failure). The cellular mechanisms underpinning these cognitive symptoms are poorly understood. Herein, the study explored the neurocognitive symptom manifestations, and tested potential therapies, in BDL mice, and used human neuronal cell cultures to explore translatability to humans. BDL animals exhibited short-term memory loss and showed reduced astrocyte coverage of the blood-brain barrier, destabilized hippocampal network activity, and neuronal senescence. Ursodeoxycholic acid (first-line therapy for most human cholestatic diseases) did not reverse symptomatic or mechanistic aspects. In contrast, obeticholic acid (OCA), a farnesoid X receptor agonist and second-line anti-cholestatic agent, normalized memory function, suppressed blood-brain barrier changes, prevented hippocampal network deficits, and reversed neuronal senescence. Co-culture of human neuronal cells with either BDL or human cholestatic patient serum induced cellular senescence and increased mitochondrial respiration, changes that were limited again by OCA. These findings provide new insights into the mechanism of cognitive symptoms in BDL animals, suggesting that OCA therapy or farnesoid X receptor agonism could be used to limit cholestasis-induced neuronal senescence.
Collapse
Affiliation(s)
- Lucy M V Gee
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ben Barron-Millar
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Claire Richardson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marco Y W Zaki
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Biochemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Saimir Luli
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel A Burgoyne
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rainie I T Cameron
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Graham R Smith
- Bioinformatics Support Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John G Brain
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Barbara Innes
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Laura Jopson
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Jessica K Dyson
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Katherine R C McKay
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexandros Pechlivanis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Elaine Holmes
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Stella Victorelli
- Department of Physiology and Biomedical Engineering, Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - George F Mells
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Richard N Sandford
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Jeremy Palmer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A Kirby
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Joao Mokochinski
- MRC London Institute of Medical Sciences, London, United Kingdom
| | - Zoe Hall
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Thomas G Bird
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom; MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Lee A Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher M Morris
- Medical Toxicology Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter S Hanson
- Medical Toxicology Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | | | - Fiona E N LeBeau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David E J Jones
- Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
15
|
Bile acids and their receptors in regulation of gut health and diseases. Prog Lipid Res 2023; 89:101210. [PMID: 36577494 DOI: 10.1016/j.plipres.2022.101210] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/26/2022]
Abstract
It is well established that bile acids play important roles in lipid metabolism. In recent decades, bile acids have also been shown to function as signaling molecules via interacting with various receptors. Bile acids circulate continuously through the enterohepatic circulation and go through microbial transformation by gut microbes, and thus bile acids metabolism has profound effects on the liver and intestinal tissues as well as the gut microbiota. Farnesoid X receptor and G protein-coupled bile acid receptor 1 are two pivotal bile acid receptors that highly expressed in the intestinal tissues, and they have emerged as pivotal regulators in bile acids metabolism, innate immunity and inflammatory responses. There is considerable interest in manipulating the metabolism of bile acids and the expression of bile acid receptors as this may be a promising strategy to regulate intestinal health and disease. This review aims to summarize the roles of bile acids and their receptors in regulation of gut health and diseases.
Collapse
|
16
|
Fan S, Feng X, Li K, Li B, Diao Y. Protective Mechanism of Ethyl Gallate against Intestinal Ischemia-Reperfusion Injury in Mice by in Vivo and in Vitro Studies Based on Transcriptomics. Chem Biodivers 2023; 20:e202200643. [PMID: 36513607 DOI: 10.1002/cbdv.202200643] [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: 07/10/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Intestinal ischemia-reperfusion injury (IIRI) is a common clinical disease that can be life-threatening in severe cases. This study aimed to investigate the effects of ethyl gallate (EG) on IIRI and its underlying mechanisms. A mouse model was established to mimic human IIRI by clamping the superior mesenteric artery. Transcriptomics techniques were used in conjunction with experiments to explore the potential mechanisms of EG action. Intestinal histomorphological damage, including intestinal villi damage and mucosal hemorrhage, was significantly reversed by EG. EG also alleviated the oxidative stress, inflammation, and intestinal epithelial apoptosis caused by IIRI. 2592 up-regulated genes and 2754 down-regulated genes were identified after EG treatment, and these differential genes were enriched in signaling pathways, including fat digestion and absorption, and extracellular matrix (ECM) receptor interactions. In IIRI mouse intestinal tissue, expression of the differential protein matrix metalloproteinase 9 (MMP9), as well as its co-protein NF-κB-p65, was significantly increased, while EG inhibited the expression of MMP9 and NF-κB-p65. In Caco-2 cells in an established oxygen-glucose deprivation/reperfusion model (OGD/R), EG significantly reversed the decrease in intestinal barrier trans-epithelial electrical resistance (TEER). However, in the presence of MMP9 inhibitors, EG did not reverse the decreasing trend in TEER. This study illustrates the protective effect and mechanism of action of EG on IIRI and, combined with in vivo and in vitro experiments, it reveals that MMP9 may be the main target of EG action. This study provides new scientific information on the therapeutic effects of EG on IIRI.
Collapse
Affiliation(s)
- Shuyuan Fan
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Xiaoyan Feng
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Kun Li
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China.,Dalian Anti-Infective Traditional Chinese Medicine, Development Engineering Technology Research Center, China
| |
Collapse
|
17
|
Fallon CM, Smyth JS, Quach A, Lajczak-McGinley N, O’Toole A, Barrett KE, Sheridan H, Keely SJ. Pentacyclic triterpenes modulate farnesoid X receptor expression in colonic epithelial cells: implications for colonic secretory function. J Biol Chem 2022; 298:102569. [PMID: 36209824 PMCID: PMC9663526 DOI: 10.1016/j.jbc.2022.102569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
The nuclear bile acid receptor, farnesoid X receptor (FXR), is an important regulator of intestinal and metabolic function. Previous studies suggest that pentacyclic triterpenes (PCTs), a class of plant-derived bioactive phytochemical, can modulate FXR activity and may therefore offer therapeutic benefits. Here, we investigated the effects of a prototypical PCT, hederagenin (HG), on FXR expression, activity, and antisecretory actions in colonic epithelial cells. T84 cells and murine enteroid-derived monolayers were employed to assess HG effects on FXR expression and activity in colonic epithelia. We measured mRNA levels by qRT-PCR and protein by ELISA and immunoblotting. Transepithelial Cl− secretion was assessed as changes in short circuit current in Ussing chambers. We determined HG treatment (5–10 μM) alone did not induce FXR activation but significantly increased expression of the receptor, both in T84 cells and murine enteroid-derived monolayers. This effect was accompanied by enhanced FXR activity, as assessed by FGF-15/19 induction in response to the synthetic, GW4064, or natural FXR agonist, chenodeoxycholic acid. Effects of HG on FXR expression and activity were mimicked by another PCT, oleanolic acid. Furthermore, we found FXR-induced downregulation of cystic fibrosis transmembrane conductance regulator Cl− channels and inhibition of transepithelial Cl− secretion were enhanced in HG-treated cells. These data demonstrate that dietary PCTs have the capacity to modulate FXR expression, activity, and antisecretory actions in colonic epithelial cells. Based on these data, we propose that plants rich in PCTs, or extracts thereof, have excellent potential for development as a new class of “FXR-targeted nutraceuticals”.
Collapse
|
18
|
Miao W, Han Y, Yang Y, Hao Z, An N, Chen J, Zhang Z, Gao X, Storey KB, Chang H, Wang S. Dynamic Changes in Colonic Structure and Protein Expression Suggest Regulatory Mechanisms of Colonic Barrier Function in Torpor-Arousal Cycles of the Daurian Ground Squirrel. Int J Mol Sci 2022; 23:ijms23169026. [PMID: 36012293 PMCID: PMC9409258 DOI: 10.3390/ijms23169026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 08/10/2022] [Indexed: 12/22/2022] Open
Abstract
Background: Both pathological conditions and hibernation can affect the barrier function of small intestine mucosa. However, the effect of hibernation on the barrier function of colonic mucosa remains unclear. Methods: We investigated morphological changes in colonic mucosa, the concentrations of specific proteins and molecules, and the enzymatic activity of diamine oxidase (DAO), in serum and colonic tissue; the expression of tight junction proteins and mucin, and the changes in inflammatory, farnesoid X receptor (FXR)–small heterodimer partner (SHP), and apoptosis-related molecules that could play a role in gut permeability changes in Daurian ground squirrels in summer active (SA), late torpor (LT), and interbout arousal (IBA) periods. Results: The results show that hibernation reduced the thickness of the colonic mucosa and the depth of the crypt, decreased the number of goblet cells (GCs), and damaged the structure of some microvilli. The concentrations of proteins and molecules, and the enzymatic activity of DAO, were all increased in the serum and colon, and the localization of tight junction proteins and mucin in the colonic mucosa were altered (compensatory response). Although the ground squirrels ate during the interbout arousal period, the changes remained similar to the response to torpor. Inflammation, apoptosis–anti-apoptosis, and FXR–SHP signaling may be involved in the possible changes in intestinal gut permeability during the torpor–arousal cycle in Daurian ground squirrels. In addition, periodic interbout arousal may play an inflammation-correcting role during the long hibernation season of Daurian ground squirrels.
Collapse
Affiliation(s)
- Weilan Miao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Yuting Han
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Yingyu Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Ziwei Hao
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Ning An
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Jiayu Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Ziwen Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Xuli Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
| | - Kenneth B. Storey
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
- Correspondence: (H.C.); (S.W.); Tel.: +86-29-88303935 (H.C.)
| | - Shiwei Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, 229# North Taibai Road, Xi’an 710069, China
- Correspondence: (H.C.); (S.W.); Tel.: +86-29-88303935 (H.C.)
| |
Collapse
|
19
|
Ocansey DKW, Zhang Z, Xu X, Liu L, Amoah S, Chen X, Wang B, Zhang X, Mao F. Mesenchymal stem cell-derived exosome mitigates colitis via the modulation of the gut metagenomics-metabolomics-farnesoid X receptor axis. Biomater Sci 2022; 10:4822-4836. [PMID: 35858469 DOI: 10.1039/d2bm00559j] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with chronic gut immune dysregulation and altered microbiome and metabolite composition. Bile acids and their receptors such as the farnesoid X receptor (FXR) form a crucial component of the chemical communications between the intestinal microbiota and the host immune system; thus, alterations in the bile acid pool affect intestinal homeostasis and exacerbate IBD. Considering the promising therapeutic effect of mesenchymal stem cell-derived exosomes (MSC-Ex) on IBD, this study assessed the regulatory effect of MSC-Ex on the gut bacteria composition and diversity, metabolites, and their related functions and pathways, as well as key inflammatory and anti-inflammatory cytokines during the mitigation of IBD. The dextran sulfate sodium (DSS)-induced IBD model of BABL/C mice was established, consisting of three groups: control, DSS, and MSC-Ex groups. Post administration of MSC-Ex, the effect was evaluated via hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), qRT-PCR, and western blotting. Mice fecal samples were obtained for metagenomics and metabolomics analysis via 16S rRNA gene sequencing and UHPLC/Q-TOF-MS respectively. Results showed that MSC-Ex mitigated colitis by significantly relieving the macroscopic and microscopic features of inflammation, modulating the gut metagenomics and metabolomics profile, and increasing colonic FXR. MSC-Ex improved the gut microbiota composition by significantly restoring the structure of OTUs and colitis-induced reduction in α-diversity, increasing the abundance of 'healthy' bacteria, decreasing disease-associated bacteria, decreasing detrimental functions, and enhancing other vital cellular functions. For the first time, we demonstrate that MSC-Ex mitigates colitis in mice by modulating the gut metagenomics-metabolomics-FXR axis, thus providing potential therapeutic targets.
Collapse
Affiliation(s)
- Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China. .,Directorate of University Health Services, University of Cape Coast, PMB, Cape Coast, Ghana
| | - Zhiyu Zhang
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212300, Jiangsu, P.R. China
| | - Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| | - Lianqin Liu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| | - Samuel Amoah
- Directorate of University Health Services, University of Cape Coast, PMB, Cape Coast, Ghana
| | - Xiang Chen
- Zhenjiang Institute for Drug and Food Control, Zhenjiang 212001, Jiangsu, P.R. China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| |
Collapse
|
20
|
Deng F, Lin ZB, Sun QS, Min Y, Zhang Y, Chen Y, Chen WT, Hu JJ, Liu KX. The role of intestinal microbiota and its metabolites in intestinal and extraintestinal organ injury induced by intestinal ischemia reperfusion injury. Int J Biol Sci 2022; 18:3981-3992. [PMID: 35844797 PMCID: PMC9274501 DOI: 10.7150/ijbs.71491] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) is a common pathophysiological process in clinical severe patients, and the effect of intestinal I/R injury on the patient's systemic pathophysiological state is far greater than that of primary intestinal injury. In recent years, more and more evidence has shown that intestinal microbiota and its metabolites play an important role in the occurrence, development, diagnosis and treatment of intestinal I/R injury. Intestinal microbiota is regulated by host genes, immune response, diet, drugs and other factors. The metabolism and immune potential of intestinal microbiota determine its important significance in host health and diseases. Therefore, targeting the intestinal microbiota and its metabolites may be an effective therapy for the treatment of intestinal I/R injury and intestinal I/R-induced extraintestinal organ injury. This review focuses on the role of intestinal microbiota and its metabolites in intestinal I/R injury and intestinal I/R-induced extraintestinal organ injury, and summarizes the latest progress in regulating intestinal microbiota to treat intestinal I/R injury and intestinal I/R-induced extraintestinal organ injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Liu H, Wang J, Ding Y, Shi X, Ren H. Antibiotic pretreatment attenuates liver ischemia-reperfusion injury by Farnesoid X receptor activation. Cell Death Dis 2022; 13:484. [PMID: 35597796 PMCID: PMC9124217 DOI: 10.1038/s41419-022-04955-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
Prophylactic antibiotics (Abx) are used before liver surgery, and the influence of antibiotic pretreatment on hepatic ischemia-reperfusion injury (IRI) remains unclear. Hence, we explored the impact of Abx pretreatment on hepatic IRI in the present work. The gut microbiota has an essential role in hepatic bile acid (BA) metabolism, and we assumed that depletion of the gut microbiota could affect the composition of hepatic BAs and affect liver IRI. The IRI model demonstrated that Abx pretreatment attenuated liver IRI by alleviating cell apoptosis, reducing the inflammatory response, and decreasing the recruitment of CCR2+ monocytes. Mechanistically, Abx pretreatment reshaped the gut microbiota, especially decreasing the relative abundance of Firmicutes and increasing the relative abundance of Clostridium, which were related to the transformation of BAs and were consistent with the altered bile acid species (unconjugated BAs, especially UDCA). These altered BAs are known FXR agonists and lead to the activation of the farnesoid X receptor (FXR), which can directly bind to the FXR response element (FXRE) harbored in the TLR4 promoter and further suppress downstream mitogen-activated protein kinase (MAPK) and nuclear kappa B (NF-κB) pathways. Meanwhile, the CCL2-CCR2 axis was also involved in the process of FXR activation, as we confirmed both in vivo and in vitro. Importantly, we proved the importance of FXR in mice and clinical occlusion samples, which were inversely correlated with liver injury. Taken together, our study identified that Abx pretreatment before liver resection was a beneficial event by activating FXR, which might become a potential therapeutic target in treating liver injury.
Collapse
Affiliation(s)
- Hanyi Liu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Institute of Hepatobiliary Surgery, Nanjing University, Nanjing, China
| | - Yitao Ding
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
- Institute of Hepatobiliary Surgery, Nanjing University, Nanjing, China.
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
- Institute of Hepatobiliary Surgery, Nanjing University, Nanjing, China.
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
- Institute of Hepatobiliary Surgery, Nanjing University, Nanjing, China.
| |
Collapse
|
22
|
Feng YD, Ye W, Tian W, Meng JR, Zhang M, Sun Y, Zhang HN, Wang SJ, Wu KH, Liu CX, Liu SY, Cao W, Li XQ. Old targets, new strategy: Apigenin-7-O-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic Biol Med 2022; 184:74-88. [PMID: 35398494 DOI: 10.1016/j.freeradbiomed.2022.03.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022]
Abstract
With the increasing morbidity and mortality, intestinal ischemia/reperfusion injury (IIRI) has attracted more and more attention, but there is no efficient therapeutics at present. Apigenin-7-O-β-D-(-6″-p-coumaroyl)-glucopyranoside (APG) is a new flavonoid glycoside isolated from Clematis tangutica that has strong antioxidant abilities in previous studies. However, the pharmacodynamic function and mechanism of APG on IIRI remain unknown. This study aimed to investigate the effects of APG on IIRI both in vivo and in vitro and identify the potential molecular mechanism. We found that APG could significantly improve intestinal edema and increase Chiu's score. MST analysis suggested that APG could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis with a dose-dependent manner. Moreover, we used siRNA silencing technology to confirm that knocking down both HO-1 and MAO-B had a positive effect on intestine. In addition, we found the HO-1 and MAO-B inhibitors also could reduce endothelial cell loss and protect vascular endothelial after reperfusion. We demonstrate that APG plays a protective role on decreasing activation of HO-1 and MAO-B, attenuating IIRI-induced ROS generation and Fe2+ accumulation, maintaining mitochondria function thus inhibiting ferroptosis.
Collapse
Affiliation(s)
- Ying-Da Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Hui-Nan Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shou-Jia Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Ke-Han Wu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chen-Xu Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shao-Yuan Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
23
|
The healing effects of Ganoderma lucidum on intestinal ischemia-reperfusion damage in rats. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1029203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
24
|
Wichman BE, Nilson J, Govindan S, Chen A, Jain A, Arun V, Derdoy J, Krebs J, Jain AK. Beyond lipids: Novel mechanisms for parenteral nutrition-associated liver disease. Nutr Clin Pract 2022; 37:265-273. [PMID: 35124837 PMCID: PMC8930621 DOI: 10.1002/ncp.10830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Parenteral nutrition (PN) is a therapy that delivers essential nutrients intravenously to patients who are unable to meet their nutrition requirements via standard enteral feeding. This methodology is often referred to as PN when accompanied by minimal or no enteral nutrition (EN). Although PN is lifesaving, significant complications can arise, such as intestinal failure-associated liver disease and gut-mucosal atrophy. The exact mechanism of injury remains ill defined. This review was designed to explore the available literature related to the drivers of injury mechanisms. The Farnesoid X receptor and fibroblast growth factor 19 signaling pathway seems to play an important role in gut-systemic signaling, and its alteration during PN provides insights into mechanistic links. Central line infections also play a key role in mediating PN-associated injury. Although lipid reduction strategies, as well as the use of multicomponent lipid emulsions and vitamin E, have shown promise, the cornerstone of preventing injury is the early establishment of EN.
Collapse
Affiliation(s)
- Brittany E Wichman
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jamie Nilson
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Srinivas Govindan
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Alan Chen
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Aditya Jain
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Varsha Arun
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Juana Derdoy
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Joseph Krebs
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ajay K Jain
- Department of Pediatrics, SSM Cardinal Glennon Hospital, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Filidou E, Tarapatzi G, Spathakis M, Papadopoulos P, Papadopoulos C, Kandilogiannakis L, Stavrou G, Doumaki E, Sioga A, Meditskou S, Arvanitidis K, Papamitsou T, Grosomanidis V, Kolios G, Kotzampassi K. Post-Cardiac Arrest Syndrome Is Not Associated With an Early Bacterial Translocation. J Surg Res 2022; 272:51-60. [PMID: 34936912 DOI: 10.1016/j.jss.2021.11.008] [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: 06/24/2021] [Revised: 10/18/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of this study was to investigate bacterial translocation and its possible role in the development of post-resuscitation inflammatory response following Cardio-Pulmonary Resuscitation (CPR) after cardiac arrest. METHODS Munich female swine were employed for a model of cardiac arrest via application of electrical current. After 7 min, CPR was initiated, and animals were either successfully return to spontaneous circulation (ROSC) within 40 min or not (no-ROSC). At the end of experimental period and prior to sacrifice, samples from the intestine, mesenteric lymph nodes (MLN), liver and portal vein blood were obtained. Evaluation of inflammation and gut permeability was performed; MLN, liver and portal vein samples were analyzed for 16 s rRNA detection and cytokine mRNA expression. RESULTS A decreased expression of the tight junction protein Occludin, with higher levels of inflammation, greater epithelial disintegration, ulceration, loss of crypts and villi height were found in the intestines of the ROSC swine in comparison to no-ROSC. The macrophage surface antigen CD-14 staining was relatively more intense in the ROSC than in no-ROSC. Higher levels of TNF-α mRNA expression were present in the liver of the ROSC group. Finally, despite the inflammatory response and the gut mucosal alterations in ROSC group, no bacterial translocation was detected in liver, MLN and portal vein. CONCLUSIONS We show that resuscitation from cardiac arrest induces inflammatory response and intestinal permeability in swine 4h after resuscitation, but not a bacterial translocation. Bacterial translocation is not an early phase phenomenon but probably part of the pathophysiologic sequelae.
Collapse
Affiliation(s)
- Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michail Spathakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Panagiotis Papadopoulos
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Charalampos Papadopoulos
- Laboratory of Biochemistry, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Stavrou
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA Hospital, University Campus, Thessaloniki, Greece; Department of Colorectal Surgery, Addenbrooke's Hospital, Cambridge, UK
| | - Eleni Doumaki
- Intensive Care Unit, 424 Military Hospital, Thessaloniki, Greece
| | - Antonia Sioga
- Laboratory of Histology- Embryology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Soultana Meditskou
- Laboratory of Histology- Embryology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Theodora Papamitsou
- Laboratory of Histology- Embryology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios Grosomanidis
- Department of Anaesthesiology, Aristotle University of Thessaloniki, AHEPA Hospital, University Campus, Thessaloniki, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA Hospital, University Campus, Thessaloniki, Greece
| |
Collapse
|
26
|
Zhang L, Li A, Huang Z, Wang Y, Yi B. Knockout of farnesoid X receptor gene aggravates cisplatin-induced kidney injury. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:174-182. [PMID: 35545407 PMCID: PMC10930519 DOI: 10.11817/j.issn.1672-7347.2022.210423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily of ligand activated transcription factors and belongs to bile acid receptor. Studies have shown that the expression of FXR in renal tissue can reduce renal injury via regulation of glucose and lipid metabolism, inhibition of inflammatory response, reduction of oxidative stress and renal fibrosis. However, it is unclear whether FXR is involved in autophagy in renal diseases. This study aims to investigate the role of FXR in cisplatin-induced acute renal injury and whether its mechanism is related to autophagy regulation. METHODS Twelve male WT or FXR-KO mice at 12 weeks were randomly divided into a WT group, a WT+cisplatin group, a FXR-KO group, and a FXR-KO+cisplatin group, with 6 mice in each group. The WT+cisplatin group and the FXR-KO+cisplatin group were intraperitoneally injected with cisplatin (20 mg/kg), and the WT group and the FXR-KO group were intraperitoneally injected with equal volume of cisplatin solvent. Seventy-two hours later, the mice were killed and blood and renal tissue samples were collected. The levels of SCr and BUN were detected by immunoturbidimetry. After the staining, the pathological changes of renal tissue were observed under optical microscope. The protein levels of LC3 and p62 were detected by Western blotting and immunohistochemistry. The clearance of damaged mitochondria and the accumulation of lysosomal substrate were observed under electron microscope. The apoptosis of renal tubular epithelial cells was detected by TUNEL. RESULTS Compared with the WT group or the FXR-KO group, both SCr and BUN levels in the WT+cisplatin group or the FXR-KO+cisplatin group were significantly increased (P<0.01 or P<0.001), and SCr and BUN levels in the FXR-KO+cisplatin group were significantly higher than those in the WT+cisplatin group (both P<0.05). Under the light microscope, there were no obvious pathological changes in the renal tissue of mice in the WT group and the FXR-KO group. Both the WT+cisplatin group and the FXR-KO+cisplatin group had vacuolar or granular degeneration of renal tubular epithelial cells, flat cells, lumen expansion, brush edge falling off, and even exposed basement membrane and tubular formation. The scores of renal tubular injury in the WT+cisplatin group and the FXR-KO+cisplatin group were significantly higher than those in the WT group and the FXR-KO group, respectively (both P<0.001), and the score in the FXR-KO+cisplatin group was significantly higher than that in the WT+cisplatin group (P<0.05). Under the transmission electron microscope, the mitochondria of mouse tubular epithelial cell in the WT+cisplatin group and the FXR-KO+cisplatin group was swollen, round, vacuolated, cristae broken or disappeared; the lysosome was uneven and high-density clumps, and the change was more obvious in the FXR-KO+cisplatin group. Western blotting showed that the ratio of LC3-II to LC3-I was decreased and the expression of p62 was increased in the WT+cisplatin group compared with the WT group and the FXR-KO+cisplatin group compared with FXR-KO group (P<0.05 or P<0.01); compared with the FXR-KO group, the ratio of LC3-II to LC3-I was decreased and the expression of p62 was increased significantly in the FXR-KO+cisplatin group (both P<0.05). Immunohistochemistry results showed that the expression of total LC3 and p62 in renal cortex of the WT+cisplatin group and the FXR-KO+cisplatin group was increased significantly, especially in the FXR-KO+cisplatin group. TUNEL results showed that the mice in the WT group and the FXR-KO group had negative staining or only a few apoptotic tubular epithelial cells, and the number of apoptotic cells in the WT+cisplatin group and the FXR-KO+cisplatin group were increased. The apoptosis rates of renal tubular epithelial cells in the WT+cisplatin group and the FXR-KO+cisplatin group were significantly higher than those in the WT group and the FXR-KO group, respectively (both P<0.001), and the apoptosis rate in the FXR-KO+cisplatin group was significantly higher than that in the WT+cisplatin group (P<0.05). CONCLUSIONS Knockout of FXR gene aggravates cisplatin induced acute renal injury, and its mechanism may be related to inhibiting autophagy and promoting apoptosis.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013.
- Medical College of Hunan Traditional Chinese Medical College, Zhuzhou Hunan 412012.
| | - Aimei Li
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Zhijun Huang
- Center of Clinical Pharmacology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yangyang Wang
- Medical College of Hunan Traditional Chinese Medical College, Zhuzhou Hunan 412012
| | - Bin Yi
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013.
| |
Collapse
|
27
|
Anderson KM, Gayer CP. The Pathophysiology of Farnesoid X Receptor (FXR) in the GI Tract: Inflammation, Barrier Function and Innate Immunity. Cells 2021; 10:cells10113206. [PMID: 34831429 PMCID: PMC8624027 DOI: 10.3390/cells10113206] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
The Farnesoid-X Receptor, FXR, is a nuclear bile acid receptor. Its originally described function is in bile acid synthesis and regulation within the liver. More recently, however, FXR has been increasingly appreciated for its breadth of function and expression across multiple organ systems, including the intestine. While FXR’s role within the liver continues to be investigated, increasing literature indicates that FXR has important roles in responding to inflammation, maintaining intestinal epithelial barrier function, and regulating immunity within the gastrointestinal (GI) tract. Given the complicated and multi-factorial nature of intestinal barrier dysfunction, it is not surprising that FXR’s role appears equally complicated and not without conflicting data in different model systems. Recent work has suggested translational applications of FXR modulation in GI pathology; however, a better understanding of FXR physiology is necessary for these treatments to gain widespread use in human disease. This review aims to discuss current scientific work on the role of FXR within the GI tract, specifically in its role in intestinal inflammation, barrier function, and immune response, while also exploring areas of controversy.
Collapse
Affiliation(s)
- Kemp M. Anderson
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Christopher P. Gayer
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, Los Angeles, CA 90027, USA
- Correspondence: ; Tel.: +1-323-361-4974
| |
Collapse
|
28
|
Dong L, Yin L, Li R, Xu L, Xu Y, Han X, Qi Y. Dioscin alleviates lung ischemia/reperfusion injury by regulating FXR-mediated oxidative stress, apoptosis, and inflammation. Eur J Pharmacol 2021; 908:174321. [PMID: 34252440 DOI: 10.1016/j.ejphar.2021.174321] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022]
Abstract
Dioscin showed various pharmacological effects in our previous studies; however, the effects and mechanisms against lung ischemia/reperfusion injury (LI/RI) have not been reported. Hypoxia/reoxygenation (H/R) models were established using A549 and primary AEC-II cells, while LI/RI models were established in rats and mice. The effects of dioscin on oxidative stress, inflammation and apoptosis in vivo and in vitro were investigated. The mechanisms were investigated focus on dioscin regulating FXR/LKB1 signaling pathway. Dioscin improved cell viability and mitochondrial membrane potential, reduced reactive oxygen species level, and inhibited H/R-mediated cell apoptosis. It also significantly decreased the lung wet/dry weight ratio, ameliorated levels of oxidative stress indicators, and enhanced the mitochondrial membrane potential and inhibited cell apoptosis in vivo. The results of mechanism research showed that dioscin activated FXR/LKB1 signals by increasing the expression of p-LKB1 and p-AMPKα, promoting the nuclear translocation of Nrf2, up-regulating the levels of HO-1, NQO1 and GCLC, expressed against oxidative stress. Furthermore, dioscin reduced Cyt C released, decreased the expression levels of Caspase-9 and Caspase-3 during apoptosis. Dioscin suppressed inflammation by inhibiting NF-κB translocation, reducing the expression levels of NF-κB, HMGB1, COX-2, IL-1β, IL-6 and TNF-α. The transfection of FXR or LKB1 siRNA further confirmed that the protective effect of dioscin against LI/RI was attributable to the regulation of FXR/LKB1 signaling pathway. Our research showed that dioscin exhibited potent activity against LI/RI, by adjusting the levels of FXR/LKB1-mediated oxidative stress, apoptosis, and inflammation, and should be considered as a new candidate for treating LI/RI.
Collapse
Affiliation(s)
- Lile Dong
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Ruomiao Li
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China.
| |
Collapse
|
29
|
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: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
30
|
Intestinal ischemic reperfusion injury: Recommended rats model and comprehensive review for protective strategies. Biomed Pharmacother 2021; 138:111482. [PMID: 33740527 DOI: 10.1016/j.biopha.2021.111482] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/01/2021] [Accepted: 03/06/2021] [Indexed: 12/17/2022] Open
Abstract
Intestinal ischemic reperfusion injury (IIRI) is a life-threatening condition with high morbidity and mortality in the clinic. IIRI was induced by intestinal ischemic diseases such as, small bowel transplantation, aortic aneurysm surgery, and strangulated hernias. Although related mechanisms have not been fully elucidated, during the last decade, researches have demonstrated that many factors are crucial in the pathological process, including oxidative stress (OS), epithelial barrier function disorder, and so on. Rats model, as the most applied animal IIRI model, provides specific targets for researches and therapeutic strategies. Moreover, various treatment strategies such as, anti-oxidative stress, anti-apoptosis, and anti-inflammation, have shown promising effects in alleviating IIRI. However, current researches cannot solve the clinical problems of IIRI, and specific treatment strategies are still needed to be exploited. This review focuses on a recommended experimental IIRI rat model and understanding of the involved mechanisms such as, OS, gut bacteria translocation, apoptosis, and necroptosis, aim at providing novel ideas for therapeutic strategies of IIRI.
Collapse
|
31
|
Xu Y, Li D, Wu J, Zhang M, Shao X, Xu L, Tang L, Zhu M, Ni Z, Zhang M, Mou S. Farnesoid X receptor promotes renal ischaemia-reperfusion injury by inducing tubular epithelial cell apoptosis. Cell Prolif 2021; 54:e13005. [PMID: 33594777 PMCID: PMC8016637 DOI: 10.1111/cpr.13005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose We investigated the role of farnesoid X receptor (FXR), a ligand‐dependent transcription factor, in renal ischaemia‐reperfusion (I/R) injury. Materials and Methods We performed unilateral renal I/R model in FXR knockout (Fxr−/−) and wild‐type (WT) mice in vivo and a hypoxia‐reoxygenation (H/R) model in vitro. The pathways by which FXR induces apoptosis were detected using a proteome profiler array. The effects of FXR on apoptosis were evaluated using immunoblotting, TUNEL assays and flow cytometry. Results Compared with WT mice, Fxr−/− mice showed improved renal function and reduced tubular injury scores and apoptosis. Consistent with the in vivo results, the silencing of FXR decreased the number of apoptotic HK‐2 cells after H/R, while FXR overexpression aggravated apoptosis. Notably, bone marrow transplantation (BMT) and immunohistochemistry experiments revealed the involvement of FXR in the tubular epithelium rather than in inflammatory cells. Furthermore, in vivo and in vitro studies demonstrated that FXR deficiency increased phosphorylated Bcl‐2 agonist of cell death (p‐Bad) expression levels and the ratio of Bcl‐2/Bcl‐xL to Bax expression in the kidney. Treatment with wortmannin, which reduced p‐Bad expression, inhibited the effects of FXR deficiency and eliminated the tolerance of Fxr−/− mouse kidneys to I/R injury. Conclusions These results established the pivotal importance of FXR inactivation in tubular epithelial cells after I/R injury. FXR may promote the apoptosis of renal tubular epithelial cells by inhibiting PI3k/Akt‐mediated Bad phosphorylation to cause renal I/R damage.
Collapse
Affiliation(s)
- Yao Xu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Dawei Li
- Department of Urology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jiajin Wu
- Department of Urology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Minfang Zhang
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xinghua Shao
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Longmei Xu
- Department of Urology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Lumin Tang
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Minyan Zhu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ming Zhang
- Department of Urology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Shan Mou
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
32
|
Zhang C, Wang Z, Feng Q, Chen WD, Wang YD. Farnesoid X receptor: a potential therapeutic target in multiple organs. Histol Histopathol 2021; 35:1403-1414. [PMID: 33393073 DOI: 10.14670/hh-18-301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Farnesoid X receptor (FXR), a member of the nuclear receptor family, is a common receptor found in the intestine and liver, and helps to maintain systemic metabolic homeostasis through regulating bile acid, glucose, lipid metabolism, and energy homeostatsis. In addition, FXR regulates the functions of various organs, such as liver, intestine, kidney, breast, pancreas, cardiovascular system and brain. FXR also plays a key role in regulation of gut-microbiota through mediating the various signaling pathways. Accordingly, FXR has become an attractive therapeutic target in a variety of diseases. This review combines classical and recent research reports to introduce the basic information about FXR and its important roles in various organs of the body.
Collapse
Affiliation(s)
- Chao Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, PR China
| | - Zixuan Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, PR China
| | - Qingqing Feng
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, PR China
| | - Wei-Dong Chen
- Key Laboratory of Molecular Pathology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China.,Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, the People's Hospital of Hebi, School of Medicine, Henan University, Henan, PR China
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, PR China.
| |
Collapse
|
33
|
Chen F, Wang D, Li X, Wang H. Molecular Mechanisms Underlying Intestinal Ischemia/Reperfusion Injury: Bioinformatics Analysis and In Vivo Validation. Med Sci Monit 2020; 26:e927476. [PMID: 33290384 PMCID: PMC7733309 DOI: 10.12659/msm.927476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a serious clinical complication. This study aimed to explore the hub genes and pathways of intestinal I/R injury. Material/Methods GSE96733 from the GEO website was extracted to analyze the differentially expressed genes (DEGs) of intestinal I/R injured and sham-operated mice at 3 h and 6 h after surgery. The DAVID and STRING databases were used to construct functional enrichment analyses of DEGs and the protein–protein interaction (PPI) network. In Cytoscape software, cytoHubba was used to identify hub genes, and MCODE was used for module analysis. Testing by qRT-PCR detected the expression of hub genes in intestinal I/R injury. Western blot analysis detected the key proteins involved with the important pathways of intestinal I/R injury. Results IL-6, IL-10, CXCL1, CXCL2, and IL-1β were identified as critical upregulated genes, while IRF7, IFIT3, IFIT1, Herc6, and Oasl2 were identified as hub genes among the downregulated genes. The qRT-PCR testing showed the expression of critical upregulated genes was significantly increased in intestinal I/R injury (P<0.05), while the expression of hub downregulated genes was notably reduced (P<0.05). The proteins of CXCL1 and CXCR2 were upregulated following intestinal I/R injury (P<0.05) and the CXCL1/CXCR2 axis was involved with intestinal I/R injury. Conclusions The results of the present study identified IL-6, IL-10, CXCL1, CXCL2, IL-1β, IRF7, IFIT3, IFIT1, Herc6, and Oasl2 as hub genes in intestinal I/R injury and identified the involvement of the CXCL1/CXCR2 axis in intestinal I/R injury.
Collapse
Affiliation(s)
- Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xiaoqian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - He Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
34
|
Ferrigno A, Palladini G, Di Pasqua LG, Berardo C, Richelmi P, Cadamuro M, Fabris L, Perlini S, Adorini L, Vairetti M. Obeticholic acid reduces biliary and hepatic matrix metalloproteinases activity in rat hepatic ischemia/reperfusion injury. PLoS One 2020; 15:e0238543. [PMID: 32911524 PMCID: PMC7482919 DOI: 10.1371/journal.pone.0238543] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background We have previously shown that obeticholic acid (OCA) upregulates the biliary excretion of asymmetric dimethylarginine (ADMA), an inhibitor of iNOS regulating the activity of matrix metalloproteinases (MMPs). Here, the effects of OCA on MMP-2 and MMP-9 activity in liver, bile and serum were evaluated after hepatic ischemia/reperfusion (I/R) injury. Material and methods Male Wistar rats (n = 20) were orally administered 10 mg/kg/day of OCA (5 days) and subjected to a 60-min ischemia and 60-min reperfusion. Bile, serum and tissue were collected for MMP-2 and MMP-9 activity quantification. The MMP regulator tissue reversion-inducing cysteine rich protein with Kazal motifs (RECK), tissue inhibitor of metalloproteinases (TIMPs), iNOS and biliary levels of LDH, γGT, glucose and ADMA were quantified. Results In the I/R group, OCA administration reduced MMP-2 and MMP-9 in liver, bile and serum. A downregulation of tissue RECK and TIMPs, observed under I/R, were recovered by OCA. Immunohistochemical staining of hepatic tissue demonstrated that RECK expression is mainly localized in both cholangiocytes and hepatocytes. Hepatic iNOS positively correlated with tissue MMP-2 and MMP-9 activity. Biliary levels of LDH, γGT and glucose were lower in I/R rats treated with OCA; in bile, MMP levels positively correlated with LDH and γGT. Conclusion Thus, OCA administration confers protection to cholangiocytes via downregulation of biliary MMPs in livers submitted to I/R. This event is associated with hepatic RECK- and TIMP-mediated MMP decrease.
Collapse
Affiliation(s)
- Andrea Ferrigno
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- * E-mail: (MV); (AF)
| | - Giuseppina Palladini
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Clarissa Berardo
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Plinio Richelmi
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | | | - Luca Fabris
- Dept. of Molecular Medicine (DMM), University of Padua, Padua, Italy
- Department of Internal Medicine, Liver Center and Section of Digestive Diseases, Yale University, New Haven, CT, United States of America
| | - Stefano Perlini
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Emergency Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luciano Adorini
- Intecept Pharmaceuticals, San Diego, CA, United States of America
| | - Mariapia Vairetti
- Dept. of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- * E-mail: (MV); (AF)
| |
Collapse
|
35
|
Perino A, Demagny H, Velazquez-Villegas L, Schoonjans K. Molecular Physiology of Bile Acid Signaling in Health, Disease, and Aging. Physiol Rev 2020; 101:683-731. [PMID: 32790577 DOI: 10.1152/physrev.00049.2019] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the past two decades, bile acids (BAs) have become established as important signaling molecules that enable fine-tuned inter-tissue communication from the liver, their site of production, over the intestine, where they are modified by the gut microbiota, to virtually any organ, where they exert their pleiotropic physiological effects. The chemical variety of BAs, to a large extent determined by the gut microbiome, also allows for a complex fine-tuning of adaptive responses in our body. This review provides an overview of the mechanisms by which BA receptors coordinate several aspects of physiology and highlights new therapeutic strategies for diseases underlying pathological BA signaling.
Collapse
Affiliation(s)
- Alessia Perino
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Hadrien Demagny
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Laura Velazquez-Villegas
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Kristina Schoonjans
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| |
Collapse
|
36
|
Jin D, Lu T, Ni M, Wang H, Zhang J, Zhong C, Shen C, Hao J, Busuttil RW, Kupiec-Weglinski JW, Zhang J, Xu N, Zhai Y. Farnesoid X Receptor Activation Protects Liver From Ischemia/Reperfusion Injury by Up-Regulating Small Heterodimer Partner in Kupffer Cells. Hepatol Commun 2020; 4:540-554. [PMID: 32258949 PMCID: PMC7109340 DOI: 10.1002/hep4.1478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Farnesoid X receptor (FXR) is the nuclear receptor of bile acids and is involved in innate immune regulation. FXR agonists have been shown to protect multiple organs from inflammatory tissue injuries. Because liver expresses high levels of FXR, we explored the potential therapeutic benefits and underlying mechanisms of pharmacologic FXR activation in a murine model of partial liver warm ischemia. Pretreatment of mice with FXR agonist 3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064) attenuated liver ischemia/reperfusion injuries (IRIs) in wild-type but not FXR knockout mice. Posttreatment with GW4064 facilitated liver recovery from IRI. Mechanistically, Kupffer cells (KCs) expressed much higher levels of FXR than bone marrow-derived macrophages (BMMs). Pretreatment of KCs but not BMMs with GW4064 resulted in lower tumor necrosis factor α but higher interleukin-10 expressions following toll-like receptor stimulation. FXR-targeted gene small heterodimer partner (SHP) was critical for the regulation of KC response by GW4064. In vivo, the depletion of KCs but not cluster of differentiation (CD) 11b+ cells or knockdown of SHP diminished the immune regulatory effect of GW4064 in liver IRI. Thus, FXR activation protects liver from IRI by up-regulating SHP in KCs to inhibit the liver proinflammatory response.
Collapse
Affiliation(s)
- Dan Jin
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA.,Department of Obstetrics and Gynecology and Shanghai Key Laboratory of Gynecologic Oncology Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Tianfei Lu
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Ming Ni
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA
| | - Han Wang
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA
| | - Jiang Zhang
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Chenpeng Zhong
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Chuan Shen
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Jun Hao
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Ronald W Busuttil
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA
| | - Jerzy W Kupiec-Weglinski
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA
| | - Jianjun Zhang
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Ning Xu
- Department of Hepatic Surgery and Liver Transplantation Center Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Yuan Zhai
- Department of Surgery David Geffen School of Medicine University of California Los Angles Los Angeles CA
| |
Collapse
|
37
|
Sun S, Zhao B, Qi M, Yao Y, Xu L, Ji R, Chen W, Wang J, Huang S, Ma L, Chen Y, Yang Z, Sheng H, Fei J, Chen E, Mao E. TUDCA Ameliorates Liver Injury Via Activation of SIRT1-FXR Signaling in a Rat Hemorrhagic Shock Model. Shock 2020; 53:217-222. [PMID: 30998645 DOI: 10.1097/shk.0000000000001351] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the changes of bile acids in the liver during hemorrhagic shock (HS) and their potential to attenuate liver injury via activation of SIRT1 (sirtuin 1)-FXR (farnesoid X receptor) signaling. METHODS A Sprague-Dawley (SD) rat HS model was established, whereas HepG2 cells were hypoxically cultured to simulate HS in vitro. Liver bile acids (BA) were profiled with ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). FXR expression was detected by western blot and immunohistochemistry. The mRNA levels of SIRT1 and FXR were detected by polymerase chain reaction. Protein expression of SIRT1, FoxM1, NF-κB, acetyl-NF-κB, p53, and acetyl-p53 was analyzed by western blot. Hepatocyte apoptosis and proliferation were measured by TUNEL assay and Ki-67 staining, respectively. Serum and supernatant cytokines were analyzed using ELISA assays. Liver injury was also assessed. To investigate the possible mechanisms, SIRT1 agonist (SRT1720), SIRT1 inhibitor (EX527), and FXR inhibitor (Z-guggulsterone) were used. RESULTS Tauroursodeoxycholic acid (TUDCA) in the liver decreased significantly after HS. SIRT1 and FXR expression was time-dependently downregulated by HS or hypoxia condition. TUDCA upregulated SIRT1-FXR activity, which inhibited expression and acetylation of NF-κB and p53 and increased FoxM1 expression, leading to decreased inflammatory response and apoptosis and increased proliferative capacity in hepatocytes, and attenuation of liver injury. EX527 pretreatment reversed the protective effect of TUDCA. Moreover, Z-guggulsterone supplementation decreased the protective effect of TUDCA in vitro. CONCLUSION TUDCA in the liver decreased during HS. TUDCA supplementation might attenuate HS-induced liver injury by upregulating SIRT1-FXR signaling.
Collapse
Affiliation(s)
- Silei Sun
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Zhao
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengzhi Qi
- Intensive Care Unit, Nanjing DrumTower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi Yao
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Xu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Ji
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlong Wang
- Emergency Centre, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shunwei Huang
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Ma
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhitao Yang
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiqiu Sheng
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Fei
- Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Prieto-Moure B, Cejalvo-Lapeña D, Belda-Antolí M, Padrón-Sanz C, Lloris-Cejalvo JM, Lloris-Carsí JM. Combination Therapy of Allopurinol and Dantrolene and Its Role In The Prevention of Experimental Ischemia Reperfusion Injury Of The Small Intestine. J INVEST SURG 2020; 34:800-807. [PMID: 31906750 DOI: 10.1080/08941939.2019.1696904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The effect of different drugs on ischemia and reperfusion (I/R; induced oxygen free radical damage) was examined in small bowel tissue because the intestine is extremely sensitive to this pathology. Different drugs (allopurinol and dantrolene) can remove oxygen free radicals or inhibit the mechanisms leading to their generation, thus reducing mucosal lesions. We investigated the protective potential of combination therapy in the intestine against I/R damage. METHODS Forty-eight male Wistar rats were separated into 8 groups: one sham (control), one I/R (ischemia 60 min + reperfusion at 24 h), and 6 groups treated with allopurinol, dantrolene, or combination therapy. The grade of injury in the small bowel was established by the lipid peroxidation (MDA) and antioxidant enzymatic activity of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) in tissue samples. Moreover, the collected samples were subjected to histological study. RESULTS Combination therapy preserved normal enzymatic levels compared to the I/R groups (p < 0.05) for all parameters studied. The animals treated with combination therapy showed less severe small bowel damage than I/R group in accordance with the histological results. CONCLUSIONS Results obtained in the experimental process indicate that the administration of antioxidants protects against intestinal damage by I/R. Overall, combination therapy may protect intestinal tissue from I/R injury.
Collapse
Affiliation(s)
- Beatriz Prieto-Moure
- Department of Biomedical Sciences, European University of Valencia, Valencia, Spain
| | - Dolores Cejalvo-Lapeña
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - Mariola Belda-Antolí
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - Carolina Padrón-Sanz
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - José Miguel Lloris-Cejalvo
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | | |
Collapse
|
39
|
Activation of the Nuclear Receptor Fxr Improves Intestinal Cell Tolerance to Ischemia-Reperfusion Injury. Shock 2019; 50:316-323. [PMID: 29028769 DOI: 10.1097/shk.0000000000001019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The farnesoid X receptor (FXR) plays an important role in bile acid metabolism, intestinal homeostasis, and intestinal ischemia-reperfusion (I/R) injury. We aimed to clarify the potential effects of FXR on intestinal epithelial cell tolerance to intestinal I/R injury and reveal the underlying mechanisms. An intestinal I/R injury model was established by the occlusion of the superior mesenteric artery for ischemia for 1 h, followed by reperfusion for 4 h in C57BL/6 (wild type [WT]) and FXR mice. The small intestine injury was assessed by histological analysis. Diamine oxidase and TNF-α levels in the serum were measured. Expressions of Bcl-2, Bax, caspase-3, and cystathionine-γ-lyase (CSE) were determined by immunohostochemical staining. Oxygen-glucose deprivation/reperfusion (OGD/R) was used to make injury in cultured Caco-2 cells pretreated with FXR agonist (INT-747) or DL-propargylglycine (PAG) for 24 h. Cell viability and the expressions of NF-κB, TNF-α, and IL-6 were assessed. Compared with WT I/R mice, FXR knockout mice exacerbated intestinal I/R injury, intestinal epithelial apoptosis, and inflammatory response. The I/R injury in WT mice was alleviated with INT-747 pretreatment. CSE expression increased after intestinal I/R injury in WT but not in FXR mice. INT-747 enhanced Caco-2 cell viability and inhibited inflammatory response by blocking the NF-κB pathway after OGD/R injury, which was diminished by a CSE-specific inhibitor (PAG). Thus, we demonstrated that FXR activation enhances intestinal epithelial cell tolerance to I/R by suppressing the inflammatory response and NF-κB pathway via CSE mediation.
Collapse
|
40
|
Dai Y, Mao Z, Han X, Xu Y, Xu L, Yin L, Qi Y, Peng J. MicroRNA-29b-3p reduces intestinal ischaemia/reperfusion injury via targeting of TNF receptor-associated factor 3. Br J Pharmacol 2019; 176:3264-3278. [PMID: 31167039 PMCID: PMC6692574 DOI: 10.1111/bph.14759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The microRNA miR-29b-3p shows important roles in regulating apoptosis and inflammation. However, its effects on intestinal ischaemia/reperfusion (II/R) injury have not been reported. Here we have investigated the functions of miR-29b-3p on II/R injury on order to find drug targets to treat the injury. EXPERIMENTAL APPROACH Two models - in vitro hypoxia/reoxygenation (H/R) of IEC-6 cells; in vivo, II/R injury in C57BL/6 mice were used. Western blotting and dual-luciferase reporter assays were used and mimic and siRNA transfection tests were applied to assess the effects of miR-29b-3p on II/R injury via targeting TNF receptor-associated factor 3 (TRAF3). KEY RESULTS The H/R procedure decreased cell viability and promoted inflammation and apoptosis in IEC-6 cells, and the II/R procedure also promoted intestinal inflammation and apoptosis in mice. Expression levels of miR-29b-3p were decreased in H/R-induced cells and II/R-induced intestinal tissues of mice compared with control group or sham group, which directly targeted TRAF3. Decreased miR-29b-3p level markedly increased TRAF3 expression via activating TGF-α-activated kinase 1 phosphorylation, increasing NF-κB (p65) levels to promote inflammation, up-regulating Bcl2-associated X expression, and down-regulating Bcl-2 expression to trigger apoptosis. In addition, the miR-29b-3p mimetic and TRAF3 siRNA in IEC-6 cells markedly suppressed apoptosis and inflammation to alleviate II/R injury via inhibiting TRAF3 signallimg. CONCLUSIONS AND IMPLICATIONS The miR-29b-3p played a critical role in II/R injury, via targeting TRAF3, which should be considered as a significant drug target to treat the disease.
Collapse
Affiliation(s)
- Yan Dai
- College of PharmacyDalian Medical UniversityDalianChina
| | - Zhang Mao
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xu Han
- College of PharmacyDalian Medical UniversityDalianChina
| | - Youwei Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lina Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lianhong Yin
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yan Qi
- College of PharmacyDalian Medical UniversityDalianChina
| | - Jinyong Peng
- College of PharmacyDalian Medical UniversityDalianChina
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning ProvinceDalian Medical UniversityDalianChina
- National‐Local Joint Engineering Research Center for Drug Development (R&D) of Neurodegenerative DiseasesDalian Medical UniversityDalianChina
| |
Collapse
|
41
|
Functional Microbiomics in Liver Transplantation: Identifying Novel Targets for Improving Allograft Outcomes. Transplantation 2019; 103:668-678. [PMID: 30507741 DOI: 10.1097/tp.0000000000002568] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gut dysbiosis, defined as a maladaptive gut microbial imbalance, has been demonstrated in patients with end-stage liver disease, defined as a contributor to disease progression, and associated clinically with severity of disease and liver-related morbidity and mortality. Despite this well-recognized phenomena in patients with end-stage liver disease, the impact of gut dysbiosis and its rate of recovery following liver transplantation (LT) remains incompletely understood. The mechanisms by which alterations in the gut microbiota impact allograft metabolism and immunity, both directly and indirectly, are multifactorial and reflect the complexity of the gut-liver axis. Importantly, while research has largely focused on quantitative and qualitative changes in gut microbial composition, changes in microbial functionality (in the presence or absence of compositional changes) are of critical importance. Therefore, to translate functional microbiomics into clinical practice, one must understand not only the compositional but also the functional changes associated with gut dysbiosis and its resolution post-LT. In this review, we will summarize critical advances in functional microbiomics in LT recipients as they apply to immune-mediated allograft injury, posttransplant complications, and disease recurrence, while highlighting potential areas for microbial-based therapeutics in LT recipients.
Collapse
|
42
|
Yan XT, Cheng XL, He XH, Zheng WZ, Xiao-Fang Y, Hu C. The HO-1-expressing bone mesenchymal stem cells protects intestine from ischemia and reperfusion injury. BMC Gastroenterol 2019; 19:124. [PMID: 31299915 PMCID: PMC6626374 DOI: 10.1186/s12876-019-1042-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 06/30/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Bone mesenchymal stromal cells (BMSC) showed protective potential against intestinal ischemia. Oxygenase-1(HO-1) could alleviate oxidative stress. In the present study, we constructed HO-1-expressing BMSC and detected the effects of it on survival, intestinal injury and inflammation following intestinal ischemia and reperfusion injury (I/R). METHODS In this experiment, eighty adult male mice were divided into Sham, I/R, I/R + BMSC, I/R + BMSC/HO-1 groups. Mice were anesthetized and intestinal I/R model were established by temporarily occluding the superior mesenteric artery for 60 min with a non-crushing clamp. Following ischemia, the clamp was removed and the intestines were allowed for reperfusion. Prior to abdominal closure, BMSC/ HO-1 (2 × 106 cells) or BMSC (2 × 106 cells) were injected into the peritoneum of I/R mice respectively. Mice were allowed to recover for 24 h and then survival rate, intestinal injury and inflammation were determined. Reactive oxygen species (ROS) was assayed by fluorescent probe. TNFα and IL-6 were assayed by ELISA. RESULTS BMSC/HO-1 increased seven day survival rate, improved intestinal injury and down-regulated inflammation after intestinal I/R when compared with sole BMSC (p < 0.05 respectively). Multiple pro-inflammatory media were also decreased following application of BMSC/HO-1, when compared with sole BMSC (p < 0.05) respectively, suggesting that BMSC /HO-1 had a better protection to intestinal I/R than BMSC therapy. CONCLUSION Administration of BMSC/HO-1 following intestinal I/R, significantly improved intestinal I/R by limiting intestinal damage and inflammation.
Collapse
Affiliation(s)
- Xue-Tao Yan
- Department of Anesthesiology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China.
| | - Xiao-Li Cheng
- Department of Anesthesiology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Xiang-Hu He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Wen-Zhong Zheng
- Department of Anesthesiology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Yuan Xiao-Fang
- Department of Anesthesiology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Chen Hu
- Department of Anesthesiology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| |
Collapse
|
43
|
Scott F, Elahi S, Adebibe M, Parampalli U, Mannur K, Góralczyk A, Sanger GJ. Farnesoid X receptor - a molecular predictor of weight loss after vertical sleeve gastrectomy? Obes Sci Pract 2019; 5:273-280. [PMID: 31275601 PMCID: PMC6587316 DOI: 10.1002/osp4.337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE To determine the expression of the bile acid receptor, farnesoid X (FXR), in human gastric mucosa and investigate correlations between expression and body-mass index (BMI) and in patients with obesity, with changes in weight and BMI following vertical sleeve gastrectomy (VSG). METHODS Human gastric mucosa was obtained from normal/overweight individuals (macroscopically-normal tissue following surgery for malignancy) or from patients with obesity (VSG). The expression of FXR and its isoforms (FXRα, FXRβ) were examined by quantitative PCR and compared with the G protein-coupled bile acid receptor, GPBA. In patients with obesity, changes in BMI and weight loss were determined following VSG. RESULTS FXRα was the predominant isoform in normal/overweight individuals. FXR expression was higher in patients with obesity but GPBA receptor expression was unchanged. For those with obesity (n = 19), no correlation was found between FXR expression and change in Body-Mass Index (BMI)/month or weight loss/month, taken 3 ± 1 months after surgery, or in BMI or weight at surgery. CONCLUSIONS Obesity is associated with increased FXR expression in the gastric mucosa. The findings are preliminary but suggest that this increase in FXR expression is a consequence of obesity, rather than its cause.
Collapse
Affiliation(s)
- F. Scott
- Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of London
| | - S. Elahi
- Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of London
| | - M. Adebibe
- Bariatric Surgery DepartmentHomerton University HospitalLondon
| | - U. Parampalli
- Bariatric Surgery DepartmentHomerton University HospitalLondon
- Royal Sussex County HospitalBrighton
| | - K. Mannur
- Bariatric Surgery DepartmentHomerton University HospitalLondon
| | - A. Góralczyk
- Bariatric Surgery DepartmentHomerton University HospitalLondon
| | - G. J. Sanger
- Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of London
| |
Collapse
|
44
|
Abstract
The term blood-bile barrier (BBlB) refers to the physical structure within a hepatic lobule that compartmentalizes and hence segregates sinusoidal blood from canalicular bile. Thus, this barrier provides physiological protection in the liver, shielding the hepatocytes from bile toxicity and restricting the mixing of blood and bile. BBlB is primarily composed of tight junctions; however, adherens junction, desmosomes, gap junctions, and hepatocyte bile transporters also contribute to the barrier function of the BBlB. Recent findings also suggest that disruption of BBlB is associated with major hepatic diseases characterized by cholestasis and aberrations in BBlB thus may be a hallmark of many chronic liver diseases. Several molecular signaling pathways have now been shown to play a role in regulating the structure and function and eventually contribute to regulation of the BBlB function within the liver. In this review, we will discuss the structure and function of the BBlB, summarize the methods to assess the integrity and function of BBlB, discuss the role of BBlB in liver pathophysiology, and finally, discuss the mechanisms of BBlB regulation. Collectively, this review will demonstrate the significance of the BBlB in both liver homeostasis and hepatic dysfunction.
Collapse
Affiliation(s)
- Tirthadipa Pradhan-Sundd
- *Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- †Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Satdarshan Pal Monga
- *Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- †Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- ‡Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Jia Q, Xie Y, Lu C, Zhang A, Lu Y, Lv S, Zhang J. Endocrine organs of cardiovascular diseases: Gut microbiota. J Cell Mol Med 2019; 23:2314-2323. [PMID: 30688023 PMCID: PMC6433674 DOI: 10.1111/jcmm.14164] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/15/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022] Open
Abstract
Gut microbiota (GM) is a collection of bacteria, fungi, archaea, viruses and protozoa, etc. They inhabit human intestines and play an essential role in human health and disease. Close information exchange between the intestinal microbes and the host performs a vital role in digestion, immune defence, nervous system regulation, especially metabolism, maintaining a delicate balance between itself and the human host. Studies have shown that the composition of GM and its metabolites are firmly related to the occurrence of various diseases. More and more researchers have demonstrated that the intestinal microbiota is a virtual 'organ' with endocrine function and the bioactive metabolites produced by it can affect the physiological role of the host. With deepening researches in recent years, clinical data indicated that the GM has a significant effect on the occurrence and development of cardiovascular diseases (CVD). This article systematically elaborated the relationship between metabolites of GM and its effects, the relationship between intestinal dysbacteriosis and cardiovascular risk factors, coronary heart disease, myocardial infarction, heart failure and hypertension and the possible pathogenic mechanisms. Regulating the GM is supposed to be a potential new therapeutic target for CVD.
Collapse
Affiliation(s)
- Qiujin Jia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingyu Xie
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunmiao Lu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ao Zhang
- Epidemiology, College of Global Public Health, New York University, New York, New York
| | - Yanmin Lu
- Tianjin Nankai Hospital, Tianjin, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
46
|
van Zutphen T, Bertolini A, de Vries HD, Bloks VW, de Boer JF, Jonker JW, Kuipers F. Potential of Intestine-Selective FXR Modulation for Treatment of Metabolic Disease. Handb Exp Pharmacol 2019; 256:207-234. [PMID: 31236687 DOI: 10.1007/164_2019_233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Farnesoid X receptor controls bile acid metabolism, both in the liver and intestine. This potent nuclear receptor not only maintains homeostasis of its own ligands, i.e., bile acids, but also regulates glucose and lipid metabolism as well as the immune system. These findings have led to substantial interest for FXR as a therapeutic target and to the recent approval of an FXR agonist for treating primary biliary cholangitis as well as ongoing clinical trials for other liver diseases. Given that FXR biology is complex, including moderate expression in tissues outside of the enterohepatic circulation, temporal expression of isoforms, posttranscriptional modifications, and the existence of several other bile acid-responsive receptors such as TGR5, clinical application of FXR modulators warrants thorough understanding of its actions. Recent findings have demonstrated remarkable physiological effects of targeting FXR specifically in the intestine (iFXR), thereby avoiding systemic release of modulators. These include local effects such as improvement of intestinal barrier function and intestinal cholesterol turnover, as well as systemic effects such as improvements in glucose homeostasis, insulin sensitivity, and nonalcoholic fatty liver disease (NAFLD). Intriguingly, metabolic improvements have been observed with both an iFXR agonist that leads to production of enteric Fgf15 and increased energy expenditure in adipose tissues and antagonists by reducing systemic ceramide levels and hepatic glucose production. Here we review the recent findings on the role of intestinal FXR and its targeting in metabolic disease.
Collapse
Affiliation(s)
- Tim van Zutphen
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- University of Groningen, Leeuwarden, The Netherlands
| | - Anna Bertolini
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Hilde D de Vries
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- University of Groningen, Leeuwarden, The Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
47
|
Bertoni S, Ballabeni V, Barocelli E, Tognolini M. Mesenteric ischemia-reperfusion: an overview of preclinical drug strategies. Drug Discov Today 2018; 23:1416-1425. [DOI: 10.1016/j.drudis.2018.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/27/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
|
48
|
Rizzetto L, Fava F, Tuohy KM, Selmi C. Connecting the immune system, systemic chronic inflammation and the gut microbiome: The role of sex. J Autoimmun 2018; 92:12-34. [PMID: 29861127 DOI: 10.1016/j.jaut.2018.05.008] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Unresolved low grade systemic inflammation represents the underlying pathological mechanism driving immune and metabolic pathways involved in autoimmune diseases (AID). Mechanistic studies in animal models of AID and observational studies in patients have found alterations in gut microbiota communities and their metabolites, suggesting a microbial contribution to the onset or progression of AID. The gut microbiota and its metabolites have been shown to influence immune functions and immune homeostasis both within the gut and systematically. Microbial derived-short chain fatty acid (SCFA) and bio-transformed bile acid (BA) have been shown to influence the immune system acting as ligands specific cell signaling receptors like GPRCs, TGR5 and FXR, or via epigenetic processes. Similarly, intestinal permeability (leaky gut) and bacterial translocation are important contributors to chronic systemic inflammation and, without repair of the intestinal barrier, might represent a continuous inflammatory stimulus capable of triggering autoimmune processes. Recent studies indicate gender-specific differences in immunity, with the gut microbiota shaping and being concomitantly shaped by the hormonal milieu governing differences between the sexes. A bi-directional cross-talk between microbiota and the endocrine system is emerging with bacteria being able to produce hormones (e.g. serotonin, dopamine and somatostatine), respond to host hormones (e.g. estrogens) and regulate host hormones' homeostasis (e.g by inhibiting gene prolactin transcription or converting glucocorticoids to androgens). We review herein how gut microbiota and its metabolites regulate immune function, intestinal permeability and possibly AID pathological processes. Further, we describe the dysbiosis within the gut microbiota observed in different AID and speculate how restoring gut microbiota composition and its regulatory metabolites by dietary intervention including prebiotics and probiotics could help in preventing or ameliorating AID. Finally, we suggest that, given consistent observations of microbiota dysbiosis associated with AID and the ability of SCFA and BA to regulate intestinal permeability and inflammation, further mechanistic studies, examining how dietary microbiota modulation can protect against AID, hold considerable potential to tackle increased incidence of AID at the population level.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy.
| | - Francesca Fava
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Kieran M Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Italy; BIOMETRA Department, University of Milan, Italy
| |
Collapse
|
49
|
Ferrigno A, Di Pasqua LG, Berardo C, Siciliano V, Rizzo V, Adorini L, Richelmi P, Vairetti M. The farnesoid X receptor agonist obeticholic acid upregulates biliary excretion of asymmetric dimethylarginine via MATE-1 during hepatic ischemia/reperfusion injury. PLoS One 2018; 13:e0191430. [PMID: 29346429 PMCID: PMC5773219 DOI: 10.1371/journal.pone.0191430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022] Open
Abstract
Background We previously showed that increased asymmetric dimethylarginine (ADMA) biliary excretion occurs during hepatic ischemia/reperfusion (I/R), prompting us to study the effects of the farnesoid X receptor (FXR) agonist obeticholic acid (OCA) on bile, serum and tissue levels of ADMA after I/R. Material and methods Male Wistar rats were orally administered 10mg/kg/day of OCA or vehicle for 5 days and were subjected to 60 min partial hepatic ischemia or sham-operated. After a 60 min reperfusion, serum, tissue and bile ADMA levels, liver mRNA and protein expression of ADMA transporters (CAT-1, CAT-2A, CAT-2B, OCT-1, MATE-1), and enzymes involved in ADMA synthesis (protein-arginine-N-methyltransferase-1, PRMT-1) and metabolism (dimethylarginine-dimethylaminohydrolase-1, DDAH-1) were measured. Results OCA administration induced a further increase in biliary ADMA levels both in sham and I/R groups, with no significant changes in hepatic ADMA content. A reduction in CAT-1, CAT-2A or CAT-2B transcripts was found in OCA-treated sham-operated rats compared with vehicle. Conversely, OCA administration did not change CAT-1, CAT-2A or CAT-2B expression, already reduced by I/R. However, a marked decrease in OCT-1 and increase in MATE-1 expression was observed. A similar trend occurred with protein expression. Conclusion The reduced mRNA expression of hepatic CAT transporters suggests that the increase in serum ADMA levels is probably due to decreased liver uptake of ADMA from the systemic circulation. Conversely, the mechanism involved in further increasing biliary ADMA levels in sham and I/R groups treated with OCA appears to be MATE-1-dependent.
Collapse
Affiliation(s)
- Andrea Ferrigno
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | | | - Clarissa Berardo
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Veronica Siciliano
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Vittoria Rizzo
- Department of Molecular Medicine, IRCCS San Matteo, University of Pavia, Pavia, Italy
| | - Luciano Adorini
- Intercept Pharmaceuticals, San Diego, California, United States of America
| | - Plinio Richelmi
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Mariapia Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- * E-mail:
| |
Collapse
|