1
|
Zhang J, Zhang X, Liu Y, Shi Y, Chen F, Leng Y. Recent insights into the effect of endoplasmic reticulum stress in the pathophysiology of intestinal ischaemia‒reperfusion injury. Biochem Biophys Res Commun 2024; 701:149612. [PMID: 38316091 DOI: 10.1016/j.bbrc.2024.149612] [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: 01/10/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Intestinal ischaemia‒reperfusion (I/R) injury is a surgical emergency. This condition is associated with a high mortality rate. At present, there are limited number of efficient therapeutic measures for this injury, and the prognosis is poor. Therefore, the pathophysiological mechanisms of intestinal I/R injury must be elucidated to develop a rapid and specific diagnostic and treatment protocol. Numerous studies have indicated the involvement of endoplasmic reticulum (ER) stress in the development of intestinal I/R injury. Specifically, the levels of unfolded and misfolded proteins in the ER lumen are increased due to unfolded protein response. However, persistent ER stress promotes apoptosis of intestinal mucosal epithelial cells through three signalling pathways in the ER, impairing intestinal mucosal barrier function and leading to the dysfunction of intestinal tissues and distant organ compartments. This review summarises the mechanisms of ER stress in intestinal I/R injury, diagnostic indicators, and related treatment strategies with the objective of providing novel insights into future therapies for this condition.
Collapse
Affiliation(s)
- Jianmin Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiaohui Zhang
- The Department of Anaesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yongqiang Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; The Department of Anaesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yajing Shi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Feng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; The Department of Anaesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
2
|
Water-Soluble Carbon Monoxide-Releasing Molecules (CORMs). Top Curr Chem (Cham) 2022; 381:3. [PMID: 36515756 DOI: 10.1007/s41061-022-00413-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/12/2022] [Indexed: 12/15/2022]
Abstract
Carbon monoxide-releasing molecules (CORMs) are promising candidates for producing carbon monoxide in the mammalian body for therapeutic purposes. At higher concentrations, CO has a harmful effect on the mammalian organism. However, lower doses at a controlled rate can provide cellular signaling for mandatory pharmacokinetic and pathological activities. To date, exploring the therapeutic implications of CO dose as a prodrug has attracted much attention due to its therapeutic significance. There are two different methods of CO insertion, i.e., indirect and direct exogenous insertion. Indirect exogenous insertion of CO suggests an advantage of reduced toxicity over direct exogenous insertion. For indirect exogenous insertion, researchers are facing the issue of tissue selectivity. To solve this issue, developers have considered the newly produced CORMs. Herein, metal carbonyl complexes (MCCs) are covalently linked with CO molecules to produce different CORMs such as CORM-1, CORM-2, and CORM-3, etc. All these CORMs required exogenous CO insertion to achieve the therapeutic targets at the optimized rate under peculiar conditions or/and triggering. Meanwhile, the metal residue was generated from i-CORMs, which can propagate toxicity. Herein, we explain CO administration, water-soluble CORMs, tissue accumulation, and cytotoxicity of depleted CORMs and the kinetic profile of CO release.
Collapse
|
3
|
Reiländer S, Schmehl W, Popp K, Nuss K, Kronen P, Verdino D, Wiezorek C, Gutmann M, Hahn L, Däubler C, Meining A, Raschig M, Kaiser F, von Rechenberg B, Scherf-Clavel O, Meinel L. Oral Use of Therapeutic Carbon Monoxide for Anyone, Anywhere, and Anytime. ACS Biomater Sci Eng 2022. [DOI: 10.1021/acsbiomaterials.2c00464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Simon Reiländer
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Wolfgang Schmehl
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Kevin Popp
- German Plastics Center (SKZ), Friedrich-Bergius-Ring 22, Wuerzburg97076, Germany
| | - Katja Nuss
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Peter Kronen
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Dagmar Verdino
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Christina Wiezorek
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Lukas Hahn
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Christof Däubler
- Department of Internal Medicine II, Gastroenterology, University Hospital Wuerzburg, Oberdürrbacherstr. 6, Wuerzburg97080, Germany
| | - Alexander Meining
- Department of Internal Medicine II, Gastroenterology, University Hospital Wuerzburg, Oberdürrbacherstr. 6, Wuerzburg97080, Germany
| | - Martina Raschig
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Friederike Kaiser
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, Würzburg97070, Germany
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty ZH, University of Zuerich, Winterthurerstrasse 260, Zuerich8057, Switzerland
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Biology (HIRI), Würzburg97070, Germany
| |
Collapse
|
4
|
Luminal administration of biliverdin ameliorates ischemia-reperfusion injury following intestinal transplant in rats. Surgery 2022; 172:1522-1528. [PMID: 36088170 DOI: 10.1016/j.surg.2022.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/23/2022] [Accepted: 07/27/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Intestinal grafts are susceptible to ischemia-reperfusion injury, resulting in the loss of mucosal barrier function and graft failure. Biliverdin is known to exert a variety of cytoprotective functions against oxidative tissue injury. Because the mucosal layer is the primary site of ischemia-reperfusion injury, mucosa-targeting strategies by luminal delivery of reagents might be beneficial. We tested whether intraluminal administration of biliverdin as an adjuvant to standard preservation solutions protected against ischemia-reperfusion injury. METHODS Orthotopic syngeneic intestinal transplants were performed on Lewis rats after 6 hours of cold preservation. Saline containing biliverdin (10 μM) or without biliverdin was introduced into the lumen of the intestinal grafts immediately before cold preservation. RESULTS Damage to the intestinal mucosa caused by ischemia-reperfusion injury resulted in severe morphological changes, including blunting of the villi and erosion, and led to significant loss of gut barrier function 3 hours after reperfusion. These changes to the mucosa were notably ameliorated by intraluminal administration of biliverdin. Biliverdin also effectively inhibited upregulation of messenger RNAs for interleukin-6, inducible nitric oxide synthase, and C-C motif chemokine 2. Additionally, biliverdin treatment prevented the loss of expression of claudin-1, a transmembrane, tight-junction barrier protein. The 14-day survival of recipients of biliverdin-treated grafts was significantly improved as compared with the recipients of saline-treated control grafts (83.3% vs 38.9%, P = .030). CONCLUSION This study demonstrated that luminally delivered biliverdin provides beneficial effects during the transplant of rat small intestinal grafts and could be an attractive therapeutic option in organ transplantation.
Collapse
|
5
|
Role of Heme Oxygenase in Gastrointestinal Epithelial Cells. Antioxidants (Basel) 2022; 11:antiox11071323. [PMID: 35883814 PMCID: PMC9311893 DOI: 10.3390/antiox11071323] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
The gastrointestinal tract is a unique organ containing both vascular and luminal routes lined by epithelial cells forming the mucosa, which play an important role in the entry of nutrients and act as a selective barrier, excluding potentially harmful agents. Mucosal surfaces establish a selective barrier between hostile external environments and the internal milieu. Heme is a major nutritional source of iron and is a pro-oxidant that causes oxidative stress. Heme oxygenases (HOs) catalyze the rate-limiting step in heme degradation, resulting in the formation of iron, carbon monoxide, and biliverdin, which are subsequently converted to bilirubin by biliverdin reductase. In gastrointestinal pathogenesis, HO-1, an inducible isoform of HO, is markedly induced in epithelial cells and plays an important role in protecting mucosal cells. Recent studies have focused on the biological effects of the products of this enzymatic reaction, which have antioxidant, anti-inflammatory, and cytoprotective functions. In this review, the essential roles of HO in the gastrointestinal tract are summarized, focusing on nutrient absorption, protection against cellular stresses, and the maintenance and regulation of tight junction proteins, emphasizing the potential therapeutic implications. The biochemical basis of the potential therapeutic implications of glutamine for HO-1 induction in gastrointestinal injury is also discussed.
Collapse
|
6
|
Obara T, Yamamoto H, Aokage T, Igawa T, Nojima T, Hirayama T, Seya M, Ishikawa-Aoyama M, Nakao A, Motterlini R, Naito H. Luminal Administration of a Water-soluble Carbon Monoxide-releasing Molecule (CORM-3) Mitigates Ischemia/Reperfusion Injury in Rats Following Intestinal Transplantation. Transplantation 2022; 106:1365-1375. [PMID: 34966108 PMCID: PMC9213078 DOI: 10.1097/tp.0000000000004007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/05/2021] [Accepted: 10/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND The protective effects of carbon monoxide (CO) against ischemia/reperfusion (IR) injury during organ transplantation have been extensively investigated. Likewise, CO-releasing molecules (CORMs) are known to exert a variety of pharmacological activities via liberation of controlled amounts of CO in organs. Therefore, we hypothesized that intraluminal administration of water-soluble CORM-3 during cold storage of intestinal grafts would provide protective effects against IR injury. METHODS Orthotopic syngeneic intestinal transplantation was performed in Lewis rats following 6 h of cold preservation in Ringer solution or University of Wisconsin solution. Saline containing CORM-3 (100 µmol/L) or its inactive counterpart (iCORM-3) was intraluminally introduced in the intestinal graft before cold preservation. RESULTS Histopathological analysis of untreated and iCORM-3-treated grafts revealed a similar erosion and blunting of the intestinal villi. These changes in the mucosa structure were significantly attenuated by intraluminal administration of CORM-3. Intestinal mucosa damage caused by IR injury led to considerable deterioration of gut barrier function 3 h postreperfusion. CORM-3 significantly inhibited upregulation of proinflammatory mRNA levels, ameliorated intestinal morphological changes, and improved graft blood flow and mucosal barrier function. Additionally, CORM-3-treated grafts increased recipient survival rates. Pharmacological blockade of soluble guanylyl cyclase activity significantly reversed the protective effects conferred by CORM-3, indicating that CO partially mediates its therapeutic actions via soluble guanylyl cyclase activation. CONCLUSIONS Our study demonstrates that luminally delivered CORM-3 provides beneficial effects in cold-stored rat small intestinal grafts and could be an attractive therapeutic application of CO in the clinical setting of organ preservation and transplantation.
Collapse
Affiliation(s)
- Takafumi Obara
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hirotsugu Yamamoto
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshiyuki Aokage
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takuro Igawa
- Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tsuyoshi Nojima
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takahiro Hirayama
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mizuki Seya
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Michiko Ishikawa-Aoyama
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Atsunori Nakao
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Hiromichi Naito
- Department of Emergency, Critical Care, and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
7
|
Role and Potential Mechanism of Heme Oxygenase-1 in Intestinal Ischemia-Reperfusion Injury. Antioxidants (Basel) 2022; 11:antiox11030559. [PMID: 35326209 PMCID: PMC8945098 DOI: 10.3390/antiox11030559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Intestinal ischemia-reperfusion (IR) injury is a complex, multifactorial, and pathophysiological condition with high morbidity and mortality, leading to serious difficulties in treatment, especially in humans. Heme oxygenase (HO) is the rate-limiting enzyme involved in heme catabolism. HO-1 (an inducible form) confers cytoprotection by inhibiting inflammation and oxidation. Furthermore, nuclear factor-erythroid 2-related factor 2 (Nrf2) positively regulates HO-1 transcription, whereas BTB and CNC homolog 1 (Bach1) competes with Nrf2 and represses its transcription. We investigated the role and potential mechanism of action of HO-1 in intestinal IR injury. Intestinal ischemia was induced for 45 min followed by 4 h of reperfusion in wild-type, Bach1-deficient, and Nrf2-deficient mice, and a carbon monoxide (CO)-releasing molecule (CORM)-3 was administered. An increase in inflammatory marker levels, nuclear factor-κB (NF-κB) activation, and morphological impairments were observed in the IR-induced intestines of wild-type mice. These inflammatory changes were significantly attenuated in Bach1-deficient mice or those treated with CORM-3, and significantly exacerbated in Nrf2-deficient mice. Treatment with an HO-1 inhibitor reversed this attenuation in IR-induced Bach1-deficient mice. Bach1 deficiency and treatment with CORM-3 resulted in the downregulation of NF-κB activation and suppression of adhesion molecules. Together, Bach1, Nrf2, and CO are valuable therapeutic targets for intestinal IR injury.
Collapse
|
8
|
Yang X, Lu W, Wang M, De La Cruz LK, Tan C, Wang B. Activated charcoal dispersion of carbon monoxide prodrugs for oral delivery of CO in a pill. Int J Pharm 2022; 618:121650. [PMID: 35276229 PMCID: PMC9060424 DOI: 10.1016/j.ijpharm.2022.121650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/12/2022]
Abstract
A novel orally bioavailable solid formulation to deliver a gaseous signaling molecule, carbon monoxide (CO), was developed by adsorbing oxalyl saccharin, a newly developed organic CO prodrug, in activated charcoal (AC). The resulting solid dispersion formulation addresses key developability issues of this CO prodrug. By taking advantage of the large surface area of AC, the paradoxical problem of low water solubility of the prodrug and the requirement of hydrolysis to release CO is resolved, and the need for an organic cosolvent is completely circumvented. The AC formulation also mitigates the adverse effect of low pH on the CO release yield, allowing steady CO release in simulated gastric and intestine fluids. This formulation allows encapsulation in normal and enteric-coated gel capsules, which enables controllable CO delivery to the upper or lower GI system. It also features an advantage of trapping CO prodrug and CO release product in the AC, therefore lowering systemic absorption of these chemicals. Through in-vivo pharmacokinetic studies in mice, the AC formulation showed better efficiency of delivering CO through oral administration compared to the prodrug dosed with an organic cosolvent. The AC formulation has also been applied to address similar developability issues of another cheletropic reaction-based CO prodrug. We envision the wide applicability of this formulation in facilitating the future development of CO-based therapeutics.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Minjia Wang
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, University, MS 38677, USA
| | - Ladie Kimberly De La Cruz
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, University, MS 38677, USA.
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
9
|
Dugbartey GJ. Emerging role of carbon monoxide in intestinal transplantation. Biomed Pharmacother 2021; 143:112237. [PMID: 34649361 DOI: 10.1016/j.biopha.2021.112237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022] Open
Abstract
Intestinal transplantation has become an established therapeutic option that provides improved quality of life to patients with end-stage intestinal failure when total parenteral nutrition fails. Whereas this challenging life-saving intervention has shown exceptional growth over the past decade, illustrating the evolution of this complex and technical procedure from its preclinical origin in the mid-20th century to become a routine clinical practice today with several recent innovations, its success is hampered by multiple hurdles including technical challenges such as surgical manipulation during intestinal graft procurement, graft preservation and reperfusion damage, resulting in poor graft quality, graft rejection, post-operative infectious complications, and ultimately negatively impacting long-term recipient survival. Therefore, strategies to improve current intestinal transplantation protocol may have a significant impact on post-transplant outcomes. Carbon monoxide (CO), previously considered solely as a toxic gas, has recently been shown to be a physiological signaling molecule at low physiological concentrations with therapeutic potentials that could overcome some of the challenges in intestinal transplantation. This review discusses recent knowledge about CO in intestinal transplantation, the underlying molecular mechanisms of protection during intestinal graft procurement, preservation, transplantation and post-transplant periods. A section of the review also discusses clinical translation of CO and its challenges in the field of solid organ transplantation.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, Ontario, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Multi-Organ Transplant Program, Western University, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana.
| |
Collapse
|
10
|
Yang X, Lu W, Wang M, Tan C, Wang B. "CO in a pill": Towards oral delivery of carbon monoxide for therapeutic applications. J Control Release 2021; 338:593-609. [PMID: 34481027 PMCID: PMC8526413 DOI: 10.1016/j.jconrel.2021.08.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023]
Abstract
Along with the impressive achievements in understanding the endogenous signaling roles and mechanism(s) of action of carbon monoxide (CO), much research has demonstrated the potential of using CO as a therapeutic agent for treating various diseases. Because of CO's toxicity at high concentrations and the observed difference in toxicity profiles of CO depending on the route of administration, this review analyzes and presents the benefits of developing orally active CO donors. Such compounds have the potential for improved safety profiles, enhancing the chance for developing CO-based therapeutics. In this review, the difference between inhalation and oral administration in terms of toxicity, CO delivery efficiency, and the potential mechanism(s) of action is analyzed. The evolution from CO gas inhalation to oral administration is also extensively analyzed by summarizing published studies up to date. The concept of "CO in a pill" can be achieved by oral administration of novel formulations of CO gas or appropriate CO donors.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wen Lu
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Minjia Wang
- Department of Pharmaceutical Sciences, University of Mississippi, MS 38677, USA
| | - Chalet Tan
- Department of Pharmaceutical Sciences, University of Mississippi, MS 38677, USA
| | - Binghe Wang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
11
|
Dugbartey GJ, Alornyo KK, Luke PPW, Sener A. Application of carbon monoxide in kidney and heart transplantation: A novel pharmacological strategy for a broader use of suboptimal renal and cardiac grafts. Pharmacol Res 2021; 173:105883. [PMID: 34525329 DOI: 10.1016/j.phrs.2021.105883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/28/2022]
Abstract
Carbon monoxide (CO) was historically regarded solely as a poisonous gas that binds to hemoglobin and reduces oxygen-carrying capacity of blood at high concentrations. However, recent findings show that it is endogenously produced in mammalian cells as a by-product of heme degradation by heme oxygenase, and has received a significant attention as a medical gas that influences a myriad of physiological and pathological processes. At low physiological concentrations, CO exhibits several therapeutic properties including antioxidant, anti-inflammatory, anti-apoptotic, anti-fibrotic, anti-thrombotic, anti-proliferative and vasodilatory properties, making it a candidate molecule that could protect organs in various pathological conditions including cold ischemia-reperfusion injury (IRI) in kidney and heart transplantation. Cold IRI is a well-recognized and complicated cascade of interconnected pathological pathways that poses a significant barrier to successful outcomes after kidney and heart transplantation. A substantial body of preclinical evidence demonstrates that CO gas and CO-releasing molecules (CO-RMs) prevent cold IRI in renal and cardiac grafts through several molecular and cellular mechanisms. In this review, we discuss recent advances in research involving the use of CO as a novel pharmacological strategy to attenuate cold IRI in preclinical models of kidney and heart transplantation through its administration to the organ donor prior to organ procurement or delivery into organ preservation solution during cold storage and to the organ recipient during reperfusion and after transplantation. We also discuss the underlying molecular mechanisms of cyto- and organ protection by CO during transplantation, and suggest its clinical use in the near future to improve long-term transplantation outcomes.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Multi-Organ Transplant Program, Western University, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Karl K Alornyo
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Patrick P W Luke
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Multi-Organ Transplant Program, Western University, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - Alp Sener
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Multi-Organ Transplant Program, Western University, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
12
|
Takagi T, Naito Y, Higashimura Y, Uchiyama K, Okayama T, Mizushima K, Katada K, Kamada K, Ishikawa T, Itoh Y. Rectal administration of carbon monoxide inhibits the development of intestinal inflammation and promotes intestinal wound healing via the activation of the Rho-kinase pathway in rats. Nitric Oxide 2021; 107:19-30. [PMID: 33340673 DOI: 10.1016/j.niox.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 11/24/2022]
Abstract
The inhalation of carbon monoxide (CO) gas and the administration of CO-releasing molecules were shown to inhibit the development of intestinal inflammation in a murine colitis model. However, it remains unclear whether CO promotes intestinal wound healing. Herein, we aimed to evaluate the therapeutic effects of the topical application of CO-saturated saline enemas on intestinal inflammation and elucidate the underlying mechanism. Acute colitis was induced with trinitrobenzene sulfonic acid (TNBS) in male Wistar rats. A CO-saturated solution was prepared via bubbling 50% CO gas into saline and was rectally administrated twice a day after colitis induction; rats were sacrificed 3 or 7 days after induction for the study of the acute or healing phases, respectively. The distal colon was isolated, and ulcerated lesions were measured. In vitro wound healing assays were also employed to determine the mechanism underlying rat intestinal epithelial cell restitution after CO treatment. CO solution rectal administration ameliorated acute TNBS-induced colonic ulceration and accelerated ulcer healing without elevating serum CO levels. The increase in thiobarbituric acid-reactive substances and myeloperoxidase activity after induction of acute TNBS colitis was also significantly inhibited after CO treatment. Moreover, the wound healing assays revealed that the CO-saturated medium enhanced rat intestinal epithelial cell migration via the activation of Rho-kinase. In addition, the activation of Rho-kinase in response to CO treatment was confirmed in the inflamed colonic tissue. Therefore, the rectal administration of a CO-saturated solution protects the intestinal mucosa from inflammation and accelerates colonic ulcer healing through enhanced epithelial cell restitution. CO may thus represent a novel therapeutic agent for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Tomohisa Takagi
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan; Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| | - Yuji Naito
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yasuki Higashimura
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, 921-8836, Japan
| | - Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Tetsuya Okayama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Katsura Mizushima
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuhiro Katada
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuhiro Kamada
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| |
Collapse
|
13
|
Peoc'h K, Puy V, Fournier T. Haem oxygenases play a pivotal role in placental physiology and pathology. Hum Reprod Update 2020; 26:634-649. [PMID: 32347305 DOI: 10.1093/humupd/dmaa014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/20/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Haem oxygenases (HO) catabolise haem, which is the prosthetic group of numerous haemoproteins. Thus, multiple primary cellular pathways and functions rely on haem availability. HO exists in two isoforms, both expressed in the placenta, namely HO-1 and HO-2, the first being inducible. Haem oxygenases, particularly HO-1, have garnered specific interest in the field of physiological and pathological placental function. These enzymes mediate haem degradation by cleaving the alpha methene bridge to produce biliverdin, which is subsequently converted to bilirubin, carbon monoxide and iron. HO-1 has anti-inflammatory and antioxidant activities. SEARCH METHODS An initial literature analysis was performed using PubMed on 3 October 2018 using key terms such as 'haem oxygenase and pregnancy', 'haem oxygenase and placenta', 'HO-1 and pregnancy', 'HO-1 and placenta', 'HO and placenta', 'HO and pregnancy', 'genetic variant and HO', 'CO and pregnancy', 'CO and placenta', 'Bilirubin and pregnancy', 'Iron and pregnancy' and 'PPAR and Haem', selecting consensus conferences, recommendations, meta-analyses, practical recommendations and reviews. A second literature analysis was performed, including notable miscarriages, foetal loss and diabetes mellitus, on 20 December 2019. The three authors studied the publications independently to decipher whether they should be included in the manuscript. OBJECTIVE AND RATIONALE This review aimed to summarise current pieces of knowledge of haem oxygenase location, function and regulation in the placenta, either in healthy pregnancies or those associated with miscarriages and foetal loss, pre-eclampsia, foetal growth restriction and diabetes mellitus. OUTCOMES HO-1 exerts some protective effects on the placentation, probably by a combination of factors, including its interrelation with the PGC-1α/PPAR pathway and the sFlt1/PlGF balance, and through its primary metabolites, notably carbon monoxide and bilirubin. Its protective role has been highlighted in numerous pregnancy conditions, including pre-eclampsia, foetal growth restriction, gestational diabetes mellitus and miscarriages. WIDER IMPLICATIONS HO-1 is a crucial enzyme in physiological and pathological placentation. This protective enzyme is currently considered a potential therapeutic target in various pregnancy diseases.
Collapse
Affiliation(s)
- Katell Peoc'h
- Université de Paris, Laboratory of Excellence GR-Ex, Centre de Recherche sur l'Inflammation, INSERM U1149, UFR de Médecine Bichat, 75018 Paris, France
- Assistance Publique des Hôpitaux de Paris, APHP Nord, Paris, France
| | - Vincent Puy
- Reproductive Biology Unit CECOS, Paris-Saclay University, Antoine Béclère Hospital, APHP, Clamart 92140, France
- Laboratoire de Développement des Gonades, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA, F-92265 Fontenay-aux-Roses, France
| | - Thierry Fournier
- Université de Paris, INSERM, UMR-S 1139, 3PHM, F-75006, Paris, France
- Fondation PremUp, F-75014, Paris, France
| |
Collapse
|
14
|
Tift MS, Alves de Souza RW, Weber J, Heinrich EC, Villafuerte FC, Malhotra A, Otterbein LE, Simonson TS. Adaptive Potential of the Heme Oxygenase/Carbon Monoxide Pathway During Hypoxia. Front Physiol 2020; 11:886. [PMID: 32792988 PMCID: PMC7387684 DOI: 10.3389/fphys.2020.00886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Heme oxygenase (HO) enzymes catalyze heme into biliverdin, releasing carbon monoxide (CO) and iron into circulation. These byproducts of heme degradation can have potent cytoprotective effects in the face of stressors such as hypoxia and ischemia-reperfusion events. The potential for exogenous use of CO as a therapeutic agent has received increasing attention throughout the past few decades. Further, HO and CO are noted as putatively adaptive in diving mammals and certain high-altitude human populations that are frequently exposed to hypoxia and/or ischemia-reperfusion events, suggesting that HO and endogenous CO afford an evolutionary advantage for hypoxia tolerance and are critical in cell survival and injury avoidance. Our goal is to describe the importance of examining HO and CO in several systems, the physiological links, and the genetic factors that underlie variation in the HO/CO pathway. Finally, we emphasize the ways in which evolutionary perspectives may enhance our understanding of the HO/CO pathway in the context of diverse clinical settings.
Collapse
Affiliation(s)
- Michael S. Tift
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Rodrigo W. Alves de Souza
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Janick Weber
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Erica C. Heinrich
- Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, United States
| | - Francisco C. Villafuerte
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, School of Medicine, San Diego, CA, United States
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, School of Medicine, San Diego, CA, United States
| |
Collapse
|
15
|
Organ preservation solutions: linking pharmacology to survival for the donor organ pathway. Curr Opin Organ Transplant 2019; 23:361-368. [PMID: 29697461 DOI: 10.1097/mot.0000000000000525] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To provide an understanding of the scientific principles, which underpinned the development of organ preservation solutions, and to bring into context new strategies and challenges for solution development against the background of changing preservation technologies and expanded criteria donor access. RECENT FINDINGS Improvements in organ preservation solutions continue to be made with new pharmacological approaches. New solutions have been developed for dynamic perfusion preservation and are now in clinical application. Principles defining organ preservation solution pharmacology are being applied for cold chain logistics in tissue engineering and regenerative medicine. SUMMARY Organ preservation solutions support the donor organ pathway. The solution compositions allow additives and pharmacological agents to be delivered direct to the target organ to mitigate preservation injury. Changing preservation strategies provide further challenges and opportunities to improve organ preservation solutions.
Collapse
|
16
|
Faizan M, Muhammad N, Niazi KUK, Hu Y, Wang Y, Wu Y, Sun H, Liu R, Dong W, Zhang W, Gao Z. CO-Releasing Materials: An Emphasis on Therapeutic Implications, as Release and Subsequent Cytotoxicity Are the Part of Therapy. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1643. [PMID: 31137526 PMCID: PMC6566563 DOI: 10.3390/ma12101643] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 02/06/2023]
Abstract
The CO-releasing materials (CORMats) are used as substances for producing CO molecules for therapeutic purposes. Carbon monoxide (CO) imparts toxic effects to biological organisms at higher concentration. If this characteristic is utilized in a controlled manner, it can act as a cell-signaling agent for important pathological and pharmacokinetic functions; hence offering many new applications and treatments. Recently, research on therapeutic applications using the CO treatment has gained much attention due to its nontoxic nature, and its injection into the human body using several conjugate systems. Mainly, there are two types of CO insertion techniques into the human body, i.e., direct and indirect CO insertion. Indirect CO insertion offers an advantage of avoiding toxicity as compared to direct CO insertion. For the indirect CO inhalation method, developers are facing certain problems, such as its inability to achieve the specific cellular targets and how to control the dosage of CO. To address these issues, researchers have adopted alternative strategies regarded as CO-releasing molecules (CORMs). CO is covalently attached with metal carbonyl complexes (MCCs), which generate various CORMs such as CORM-1, CORM-2, CORM-3, ALF492, CORM-A1 and ALF186. When these molecules are inserted into the human body, CO is released from these compounds at a controlled rate under certain conditions or/and triggers. Such reactions are helpful in achieving cellular level targets with a controlled release of the CO amount. However on the other hand, CORMs also produce a metal residue (termed as i-CORMs) upon degradation that can initiate harmful toxic activity inside the body. To improve the performance of the CO precursor with the restricted development of i-CORMs, several new CORMats have been developed such as micellization, peptide, vitamins, MOFs, polymerization, nanoparticles, protein, metallodendrimer, nanosheet and nanodiamond, etc. In this review article, we shall describe modern ways of CO administration; focusing primarily on exclusive features of CORM's tissue accumulations and their toxicities. This report also elaborates on the kinetic profile of the CO gas. The comprehension of developmental phases of CORMats shall be useful for exploring the ideal CO therapeutic drugs in the future of medical sciences.
Collapse
Affiliation(s)
- Muhammad Faizan
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Niaz Muhammad
- Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China.
| | | | - Yongxia Hu
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Yanyan Wang
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Ya Wu
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Huaming Sun
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Ruixia Liu
- Institute of Process Engineering, Chinese Academy of Science, Beijing 100190, China.
| | - Wensheng Dong
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Weiqiang Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| | - Ziwei Gao
- Key Laboratory of Applied Surface and Colloid Chemistry MOE, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
17
|
Weissenbacher A, Vrakas G, Nasralla D, Ceresa CDL. The future of organ perfusion and re-conditioning. Transpl Int 2019; 32:586-597. [PMID: 30980772 PMCID: PMC6850430 DOI: 10.1111/tri.13441] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/25/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
Organ preservation and re‐conditioning using machine perfusion technologies continue to generate promising results in terms of viability assessment, organ utilization and improved initial graft function. Here, we summarize the latest findings and study the results of ex‐vivo/ex‐situ hypothermic (HMP) and normothermic machine perfusion (NMP) in the area of abdominal organ transplantation (kidney, liver, pancreas and intestine). We also consider the potential role of normothermic regional perfusion (NRP) to re‐condition donors after circulatory death organs before retrieval. The findings from clinical studies reported to date suggest that machine perfusion will offer real benefits when compared with conventional cold preservation. Several randomized trials are expected to report their findings within the next 2 years which may shed light on the relative merits of different perfusion methods and could indicate which perfusion parameters may be most useful to predict organ quality and viability. Further work is needed to identify composite endpoints that are relevant for transplanted organs that have undergone machine preservation. Multi‐centre trials to compare and analyse the combinations of NRP followed by HMP and/or NMP, either directly after organ retrieval using transportable devices or when back‐to‐base, are needed. The potential applications of machine preservation technology beyond the field of solid organ transplantation are also considered.
Collapse
Affiliation(s)
- Annemarie Weissenbacher
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria.,Nuffield Department of Surgical Sciences, Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Georgios Vrakas
- Nuffield Department of Surgical Sciences, Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - David Nasralla
- Nuffield Department of Surgical Sciences, Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Carlo D L Ceresa
- Nuffield Department of Surgical Sciences, Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Liu S, Chen HZ, Xu ZD, Wang F, Fang H, Bellanfante O, Chen XL. Sodium butyrate inhibits the production of HMGB1 and attenuates severe burn plus delayed resuscitation-induced intestine injury via the p38 signaling pathway. Burns 2018; 45:649-658. [PMID: 30482615 DOI: 10.1016/j.burns.2018.09.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inflammatory response triggered by high mobility group box-1 (HMGB1) protein and oxidative stress play critical roles in the intestinal injury after severe burn. Sodium butyrate, a histone deacetylase inhibitor, has potential anti-inflammatory properties, inhibiting the expression of inflammatory mediators such as HMGB1 in diverse diseases. This study was designed to investigate the effects of sodium butyrate on severe burn plus delayed resuscitation-induced intestine injury, intestinal expressions of HMGB1 and intracellular adhesion molecule-1 (ICAM-1), oxidative stress, and signal transduction pathway changes in rats. MATERIALS AND METHODS Fifty-six Sprague-Dawley rats were divided into 3 groups randomly: (1) sham group, animals underwent sham burn; (2) burn group, rats subjected to full-thickness burns of 30% total body surface area (TBSA) and received 2ml/kg/TBSA lactated Ringer solution for resuscitation at 6, 12, and 36h after burn injury; (3) burn plus sodium butyrate (burn+SB) group, animals received burn injury and lactated Ringer solution with sodium butyrate inside for resuscitation in the same manner. Diamine oxidase (DAO) concentration in plasma was measured by enzyme-linked immunosorbent assay. Intestinal fatty acid binding protein (I-FABP) and ICAM-1 expressions in the intestine were analyzed by immunohistochemical method. HMGB1 and p38 mitogen-activated protein kinase (MAPK) expressions in the intestine tissues were examined by Western blot. The intestinal concentration of malondialdehyde (MDA) was also determined. RESULTS Intestinal HMGB1 expression was significantly increased in burn group compared with sham group. Sodium butyrate administration significantly inhibited the HMGB1 expression in the intestine, decreased the DAO concentration in plasma, reduced the intestinal I-FABP expression, and improved the intestinal histologic changes induced by burn injury plus delayed resuscitation. Sodium butyrate treatment also markedly reduced the increase of intestinal ICAM-1 expression and MDA content, and inhibited p38 MAPK activity in the intestine of severely burned rats with delayed resuscitation. CONCLUSIONS Sodium butyrate inhibits HMGB1 expression which could be attributed to p38 MAPK signal transduction pathway and decreases intestinal inflammatory responses and oxidative stress, thus attenuates burn plus delayed resuscitation-induced intestine injury.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Hong-Ze Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Zheng-Dong Xu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Fei Wang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, PR China
| | - Ophelia Bellanfante
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China.
| |
Collapse
|
19
|
De Schepper S, Stakenborg N, Matteoli G, Verheijden S, Boeckxstaens GE. Muscularis macrophages: Key players in intestinal homeostasis and disease. Cell Immunol 2017; 330:142-150. [PMID: 29291892 PMCID: PMC6108422 DOI: 10.1016/j.cellimm.2017.12.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022]
Abstract
Muscularis macrophages densily colonize the outermost layer of the gastrointestinal tract. Muscularis macrophages communicate with enteric neurons in a bidirectional matter. Muscularis macrophages are tissue-protective but can contribute to disease. Current challenges are to decipher therapeutic potentials of muscularis macrophages.
Macrophages residing in the muscularis externa of the gastrointestinal tract are highly specialized cells that are essential for tissue homeostasis during steady-state conditions as well as during disease. They are characterized by their unique protective functional phenotype that is undoubtedly a consequence of the reciprocal interaction with their environment, including the enteric nervous system. This muscularis macrophage-neuron interaction dictates intestinal motility and promotes tissue-protection during injury and infection, but can also contribute to tissue damage in gastrointestinal disorders such as post-operative ileus and gastroparesis. Although the importance of muscularis macrophages is clearly recognized, different aspects of these cells remain largely unexplored such their origin, longevity and instructive signals that determine their function and phenotype. In this review, we will discuss the phenotype, functions and origin of muscularis macrophages during steady-state and disease conditions. We will highlight the bidirectional crosstalk with neurons and potential therapeutic strategies that target and manipulate muscularis macrophages to restore their protective signature as a treatment for disease.
Collapse
Affiliation(s)
- Sebastiaan De Schepper
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Nathalie Stakenborg
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Laboratory for Mucosal Immunology, University of Leuven, Leuven, Belgium.
| | - Simon Verheijden
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Guy E Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Ling K, Men F, Wang WC, Zhou YQ, Zhang HW, Ye DW. Carbon Monoxide and Its Controlled Release: Therapeutic Application, Detection, and Development of Carbon Monoxide Releasing Molecules (CORMs). J Med Chem 2017; 61:2611-2635. [PMID: 28876065 DOI: 10.1021/acs.jmedchem.6b01153] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carbon monoxide (CO) is attracting increasing attention because of its role as a gasotransmitter with cytoprotective and homeostatic properties. Carbon monoxide releasing molecules (CORMs) are spatially and temporally controlled CO releasers that exhibit superior and more effective pharmaceutical traits than gaseous CO because of their chemistry and structure. Experimental and preclinical research in animal models has shown the therapeutic potential of inhaled CO and CORMs, and the biological effects of CO and CORMs have also been observed in preclinical trials via the genetic modulation of heme oxygenase-1 (HO-1). In this review, we describe the pharmaceutical use of CO and CORMs, methods of detecting CO release, and developments in CORM design and synthesis. Many valuable clinical CORMs formulated using macromolecules and nanomaterials are also described.
Collapse
Affiliation(s)
- Ken Ling
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China.,Department of Anesthesiology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Fang Men
- College of Chemistry and Molecular Sciences , Wuhan University , Wuhan 430072 , China
| | - Wei-Ci Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Hao-Wen Zhang
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| |
Collapse
|
21
|
Riella LV, Bagley J, Iacomini J, Alegre ML. Impact of environmental factors on alloimmunity and transplant fate. J Clin Invest 2017; 127:2482-2491. [PMID: 28481225 DOI: 10.1172/jci90596] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although gene-environment interactions have been investigated for many years to understand people's susceptibility to autoimmune diseases or cancer, a role for environmental factors in modulating alloimmune responses and transplant outcomes is only now beginning to emerge. New data suggest that diet, hyperlipidemia, pollutants, commensal microbes, and pathogenic infections can all affect T cell activation, differentiation, and the kinetics of graft rejection. These observations reveal opportunities for novel therapeutic interventions to improve graft outcomes as well as for noninvasive biomarker discovery to predict or diagnose graft deterioration before it becomes irreversible. In this Review, we will focus on the impact of these environmental factors on immune function and, when known, on alloimmune function, as well as on transplant fate.
Collapse
Affiliation(s)
- Leonardo V Riella
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jessamyn Bagley
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Boston, Massachusetts, USA
| | - John Iacomini
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Boston, Massachusetts, USA
| | | |
Collapse
|
22
|
Oltean M, Jiga L, Hellström M, Söfteland J, Papurica M, Hoinoiu T, Ionac M, Casselbrant A. A sequential assessment of the preservation injury in porcine intestines. J Surg Res 2017; 216:149-157. [PMID: 28807200 DOI: 10.1016/j.jss.2017.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/20/2017] [Accepted: 05/01/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Clinical and experimental evidence strongly suggest that ischemia-reperfusion injury after intestinal transplantation has deleterious short- and long-term effects and finding means to reduce ischemia-reperfusion injury is a major research area. The anatomical and physiological similarities between the human and porcine digestive tract favor its use as a preclinical model for translational research. Intriguingly, no systematic appraisal of the development of the intestinal preservation injury in pigs is available. MATERIALS AND METHODS Intestinal procurement was performed in nine pigs using histidine-tryptophan-ketoglutarate solution as preservation fluid. Ileal biopsies were obtained after 8, 14, and 24 h of static cold storage (SCS), and the preservation injury was assessed morphologically (Chiu score) as well as on the molecular level. Tight junction (zonula occludens, claudin-3 and 4, tricellulin, and occludin) and adherens junctions (E-cadherin) proteins were studied using immunofluorescence and Western blot. RESULTS Eight hours of SCS induced minimal mucosal changes (Chiu grade 1) that advanced to significant subepithelial edema (Chiu grade 3) after 24 h; progressive Goblet cell depletion was also noted. Apoptosis (studied by cleaved caspase-3 staining significantly increased after 24 h of SCS. Significant molecular changes with decreasing expression of zonula occludens, tricellulin, and occludin were evident already after 8 h of SCS and continuously worsened. Claudin-3 and Claudin-4 and E-cadherin expression remained relatively unaltered during SCS. CONCLUSIONS Important molecular alterations precede histologic changes during SCS of the porcine intestine and may be used as more sensitive injury markers than histologic changes in intestinal ischemia and transplantation.
Collapse
Affiliation(s)
- Mihai Oltean
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, Timisoara, Romania; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Lucian Jiga
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, Timisoara, Romania; Department for Plastic, Reconstructive, Aesthetic and Hand Surgery, Evangelisches Krankenhaus, Medical Campus, University of Oldenburg, Oldenburg, Germany
| | - Mats Hellström
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - John Söfteland
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marius Papurica
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, Timisoara, Romania
| | - Teodora Hoinoiu
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, Timisoara, Romania
| | - Mihai Ionac
- Pius Branzeu Center for Laparoscopic Surgery and Microsurgery, University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna Casselbrant
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Steiger C, Hermann C, Meinel L. Localized delivery of carbon monoxide. Eur J Pharm Biopharm 2016; 118:3-12. [PMID: 27836646 DOI: 10.1016/j.ejpb.2016.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/18/2016] [Accepted: 11/07/2016] [Indexed: 01/18/2023]
Abstract
The heme oxygenase (HO)/carbon monoxide (CO) system is a physiological feedback loop orchestrating various cell-protective effects in response to cellular stress. The therapeutic use of CO is impeded by safety challenges as a result of high CO-Hemoglobin formation following non-targeted, systemic administration jeopardizing successful CO therapies as of this biological barrier. Another caveat is the use of CO-Releasing Molecules containing toxicologically critical transition metals. An emerging number of local delivery approaches addressing these issues have recently been introduced and provide exciting new starting points for translating the fascinating preclinical potential of CO into a clinical setting. This review will discuss these approaches and link to future delivery strategies aiming at establishing CO as a safe and effective medication of tomorrow.
Collapse
Affiliation(s)
- Christoph Steiger
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Cornelius Hermann
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany.
| |
Collapse
|
24
|
Abstract
Carbon monoxide (CO) has long been considered an environmental pollutant and a poison. Exogenous exposure to amounts of CO beyond the physiologic level of the body can result in a protective or adaptive response. However, as a gasotransmitter, endogenous CO is important for multiple physiologic functions. To date, at least seven distinct methods of delivering CO have been utilized in animal and clinical studies. In this mini-review, we summarize the exogenous CO delivery methods and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Hui-Jun Hu
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China; Department of Hyperbaric Oxygen, Navy General Hospital, Beijing, China
| | - Qiang Sun
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing, China
| | - Zhou-Heng Ye
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xue-Jun Sun
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Takagi T, Naito Y, Uchiyama K, Mizuhima K, Suzuki T, Horie R, Hirata I, Tsuboi H, Yoshikawa T. Carbon monoxide promotes gastric wound healing in mice via the protein kinase C pathway. Free Radic Res 2016; 50:1098-1105. [DOI: 10.1080/10715762.2016.1189546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizuhima
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Suzuki
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryusuke Horie
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ikuhiro Hirata
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisato Tsuboi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshikazu Yoshikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
26
|
Ryter SW, Choi AMK. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl Res 2016; 167:7-34. [PMID: 26166253 PMCID: PMC4857893 DOI: 10.1016/j.trsl.2015.06.011] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022]
Abstract
The heme oxygenase-1 (HO-1) enzyme system remains an attractive therapeutic target for the treatment of inflammatory conditions. HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXα (BV), the latter which is converted to bilirubin-IXα (BR). HO-1 may function as a pleiotropic regulator of inflammatory signaling programs through the generation of its biologically active end products, namely CO, BV and BR. CO, when applied exogenously, can affect apoptotic, proliferative, and inflammatory cellular programs. Specifically, CO can modulate the production of proinflammatory or anti-inflammatory cytokines and mediators. HO-1 and CO may also have immunomodulatory effects with respect to regulating the functions of antigen-presenting cells, dendritic cells, and regulatory T cells. Therapeutic strategies to modulate HO-1 in disease include the application of natural-inducing compounds and gene therapy approaches for the targeted genetic overexpression or knockdown of HO-1. Several compounds have been used therapeutically to inhibit HO activity, including competitive inhibitors of the metalloporphyrin series or noncompetitive isoform-selective derivatives of imidazole-dioxolanes. The end products of HO activity, CO, BV and BR may be used therapeutically as pharmacologic treatments. CO may be applied by inhalation or through the use of CO-releasing molecules. This review will discuss HO-1 as a therapeutic target in diseases involving inflammation, including lung and vascular injury, sepsis, ischemia-reperfusion injury, and transplant rejection.
Collapse
Affiliation(s)
- Stefan W Ryter
- Joan and Sanford I. Weill Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY.
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY
| |
Collapse
|
27
|
Steiger C, Wollborn J, Gutmann M, Zehe M, Wunder C, Meinel L. Controlled therapeutic gas delivery systems for quality-improved transplants. Eur J Pharm Biopharm 2015; 97:96-106. [PMID: 26527426 DOI: 10.1016/j.ejpb.2015.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/16/2015] [Accepted: 10/18/2015] [Indexed: 12/15/2022]
Abstract
Therapeutic gases enriched into perfusion solutions have been effectively used for the improvement of organ transplant quality. At present, the enrichment of perfusion solutions with gases requires complex machinery/containers and handling precautions. Alternatively, the gas is generated within the perfusion solution by supplemented carbonylated transition metal complexes with associated toxicological concerns when these metals contact the transplant. Therefore, we developed therapeutic gas releasing systems (TGRSs) allowing for the controlled generation and release of therapeutic gases (carbon monoxide and hydrogen sulfide) from otherwise hermetically sealed containers, such that the perfusion solution for the transplant is saturated with the gas but no other components from the TGRS are liberated in the solution. The release from the TGRS into the perfusion solution can be tailored as a function of the number and thickness of gas permeable membranes leading to release patterns having been linked to therapeutic success in previous trials. Furthermore, the surrogate biomarker HMGB1 was significantly downregulated in ischemic rat liver transplants perfused with enriched CO solution as compared to control. In conclusion, the TGRS allows for easy, reliable, and controlled generation and release of therapeutic gases while removing safety concerns of current approaches, thereby positively impacting the risk benefit profile of using therapeutic gases for transplant quality improvement in the future.
Collapse
Affiliation(s)
- Christoph Steiger
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Jakob Wollborn
- Department of Anaesthesia and Critical Care, University of Wuerzburg, Oberduerrbacherstraße 6, DE-97080 Wurzburg, Germany; Department of Anesthesiology and Intensive Care Medicine, University Medical Center Freiburg, Hugstetter Str. 55, DE-79106 Freiburg, Germany
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Markus Zehe
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Christian Wunder
- Department of Anaesthesia and Critical Care, University of Wuerzburg, Oberduerrbacherstraße 6, DE-97080 Wurzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany.
| |
Collapse
|
28
|
Intestinal preservation for transplantation: current status and alternatives for the future. Curr Opin Organ Transplant 2015; 20:308-13. [PMID: 25944227 DOI: 10.1097/mot.0000000000000187] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Among transplantable abdominal organs the intestine has the shortest cold storage time, raising significant medical and logistical challenges. Herein, established and innovative, emerging concepts in intestinal preservation are summarized. RECENT FINDINGS The method of intestinal preservation using an in-situ vascular perfusion followed by static storage remained unchanged for almost 30 years, despite suboptimal results. Advanced preservation injury occurs within 12 h and is little influenced by the type of solution used. Recent reports indicate that several customized luminal solutions containing various amino acids and macromolecules may delay its development. In addition, gaseous interventions in the storage solutions or in the lumen seem promising and easily applicable tools that may further reduce the ischemia-reperfusion injury and safely prolong the preservation time. Rodent models are not entirely suitable for direct translation to clinical practice as the development of preservation injury is faster than in humans. SUMMARY The limitations of intestinal preservation originate in the methods (vascular perfusion and static storage) rather than in the solutions used. Several additional strategies promise to prolong the cold storage and reduce its impact on the intestinal graft and deserve further exploration in large animals and clinical studies.
Collapse
|
29
|
Katada K, Takagi T, Uchiyama K, Naito Y. Therapeutic roles of carbon monoxide in intestinal ischemia-reperfusion injury. J Gastroenterol Hepatol 2015; 30 Suppl 1:46-52. [PMID: 25827804 DOI: 10.1111/jgh.12742] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal ischemia-reperfusion (I-R) injury is a complex, multifactorial, pathophysiological process with high morbidity and mortality, leading to serious difficulty in treatment. The mechanisms involved in the pathogenesis of intestinal I-R injury have been examined in detail and various therapeutic approaches for intestinal I-R injury have been developed; however, existing circumstances have not yet led to a dramatic change of treatment. Carbon monoxide (CO), one of the by-products of heme degradation by heme oxygenase (HO), is considered as a candidate for treatment of intestinal I-R injury and indeed HO-1-derived endogenous CO and exogenous CO play a pivotal role in protecting the gastrointestinal tract from intestinal I-R injury. Interestingly, anti-inflammatory effects of CO have been elucidated sufficiently in various cell types including endothelial cells, circulating leukocytes, macrophages, lymphocytes, epithelial cells, fibroblast, organ-specific cells, and immune-presenting cells. In this review, we herein focus on the therapeutic roles of CO in intestinal I-R injury and the cell-specific anti-inflammatory effects of CO, clearly demonstrating future therapeutic strategies of CO for treating intestine I-R injury.
Collapse
Affiliation(s)
- Kazuhiro Katada
- Molecular Gastroenterology and Hepatology, Graduate School of Medial Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | |
Collapse
|
30
|
Babu D, Motterlini R, Lefebvre RA. CO and CO-releasing molecules (CO-RMs) in acute gastrointestinal inflammation. Br J Pharmacol 2014; 172:1557-73. [PMID: 24641722 DOI: 10.1111/bph.12632] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 12/13/2022] Open
Abstract
Carbon monoxide (CO) is enzymatically generated in mammalian cells alongside the liberation of iron and the production of biliverdin and bilirubin. This occurs during the degradation of haem by haem oxygenase (HO) enzymes, a class of ubiquitous proteins consisting of constitutive and inducible isoforms. The constitutive HO2 is present in the gastrointestinal tract in neurons and interstitial cells of Cajal and CO released from these cells might contribute to intestinal inhibitory neurotransmission and/or to the control of intestinal smooth muscle cell membrane potential. On the other hand, increased expression of the inducible HO1 is now recognized as a beneficial response to oxidative stress and inflammation. Among the products of haem metabolism, CO appears to contribute primarily to the antioxidant and anti-inflammatory effects of the HO1 pathway explaining the studies conducted to exploit CO as a possible therapeutic agent. This article reviews the effects and, as far as known today, the mechanism(s) of action of CO administered either as CO gas or via CO-releasing molecules in acute gastrointestinal inflammation. We provide here a comprehensive overview on the effect of CO in experimental in vivo models of post-operative ileus, intestinal injury during sepsis and necrotizing enterocolitis. In addition, we will analyse the in vitro data obtained so far on the effect of CO on intestinal epithelial cell lines exposed to cytokines, considering the important role of the intestinal mucosa in the pathology of gastrointestinal inflammation.
Collapse
Affiliation(s)
- D Babu
- Heymans Institute of Pharmacology, Ghent University, Gent, Belgium
| | | | | |
Collapse
|
31
|
Abstract
SIGNIFICANCE Heme oxygenase enzymes, which exist as constitutive (HO-2) and inducible (HO-1) isoforms, degrade heme to carbon monoxide (CO) and the bile pigment biliverdin. In the last two decades, substantial scientific evidence has been collected on the function of HO-1 in cell homeostasis, emphasizing these two important features: (i) HO-1 is a fundamental "sensor" of cellular stress and directly contributes toward limiting or preventing tissue damage; (ii) the products of HO-1 activity dynamically participate in cellular adaptation to stress and are inherently involved in the mechanisms of defence. RECENT ADVANCES On the basis of its promising cytoprotective features, scientists have pursued the targeting of HO-1 as an attractive cellular pathway for drug discovery. Three different pharmacological approaches are currently being investigated in relation to HO-1, namely the use of CO gas, the development of CO-releasing molecules (CO-RMs), and small molecules possessing the ability to up-regulate HO-1 in cells and tissues. CRITICAL ISSUE: Studies on the regulation and amplification of the HO-1/CO pathway by selective pharmacological approaches may lead to the discovery of novel drugs for the treatment of a variety of diseases. FUTURE DIRECTIONS In this review, we will discuss in detail the importance of pharmacologically manipulating the HO-1 pathway and its products for conferring protection against a variety of conditions that are characterized by oxidative stress and inflammation. We will also evaluate each of the strategic approaches being developed by considering their intrinsic advantages and disadvantages, which may have implications for their use as therapeutics in specific pathological conditions.
Collapse
|
32
|
Romanski S, Stamellou E, Jaraba JT, Storz D, Krämer BK, Hafner M, Amslinger S, Schmalz HG, Yard BA. Enzyme-triggered CO-releasing molecules (ET-CORMs): evaluation of biological activity in relation to their structure. Free Radic Biol Med 2013; 65:78-88. [PMID: 23774042 DOI: 10.1016/j.freeradbiomed.2013.06.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/08/2013] [Accepted: 06/07/2013] [Indexed: 10/26/2022]
Abstract
Acyloxydiene-Fe(CO)3 complexes act as enzyme-triggered CO-releasing molecules (ET-CORMs) and can deliver CO intracellularly via esterase-mediated hydrolysis. The protective properties of structurally different ET-CORMs on hypothermic preservation damage and their ability to inhibit VCAM-1 expression were tested on cultured human umbilical vein endothelial cells (HUVEC) and renal proximal tubular epithelial cells (PTEC) using a structure-activity approach. Cytotoxicity of ET-CORMs, protection against hypothermic preservation damage, and inhibition of VCAM-1 expression were assessed. Cytotoxicity of 2-cyclohexenone and 1,3-cyclohexanedione-derived ET-CORMs was more pronounced in HUVEC compared to PTEC and was dependent on the position and type of the ester (acyloxy) substituent(s) (acetate>pivalate>palmitate). Protection against hypothermic preservation injury was only observed for 2-cyclohexenone-derived ET-CORMs and was not mediated by the ET-CORM decomposition product 2-cyclohexenone itself. Structural requirements for protection by these ET-CORMs were different for HUVEC and PTEC. Protection was affected by the nature of the ester functionality in both cell lines. VCAM-1 expression was inhibited by both 2-cyclohexenone- and 1,3-cyclohexanedione-derived ET-CORMs. 2-Cyclohexenone, but not 1,3-cyclohexanedione, also inhibited VCAM-1 expression. We demonstrate that structural alterations of ET-CORMs significantly affect their biological activity. Our data also indicate that different ET-CORMs behave differently in various cell types (epithelial vs endothelial). These findings warrant further studies not only to elucidate the structure-activity relation of ET-CORMs in mechanistic terms but also to assess if structural optimization will yield ET-CORMs with restricted cell specificity.
Collapse
Affiliation(s)
- S Romanski
- Department of Chemistry, University of Cologne, Greinstr. 4, 50939 Köln, Germany
| | - E Stamellou
- Vth Medical Department, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - J T Jaraba
- Vth Medical Department, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - D Storz
- Vth Medical Department, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - B K Krämer
- Vth Medical Department, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - M Hafner
- Institut für Molekülar- and Zellbiologie, Hochschule Mannheim, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - S Amslinger
- Institut für Organische Chemie, Universität Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - H G Schmalz
- Department of Chemistry, University of Cologne, Greinstr. 4, 50939 Köln, Germany
| | - B A Yard
- Vth Medical Department, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
33
|
Gibbons SJ, Verhulst PJ, Bharucha A, Farrugia G. Review article: carbon monoxide in gastrointestinal physiology and its potential in therapeutics. Aliment Pharmacol Ther 2013; 38:689-702. [PMID: 23992228 PMCID: PMC3788684 DOI: 10.1111/apt.12467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/03/2013] [Accepted: 08/07/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND While carbon monoxide (CO) is a known toxin, it is now recognised that CO is also an important signalling molecule involved in physiology and pathophysiology. AIMS To summarise our current understanding of the role of endogenous CO in the regulation of gastrointestinal physiology and pathophysiology, and to potential therapeutic applications of modulating CO. METHODS This review is based on a comprehensive search of the Ovid Medline comprehensive database and supplemented by our ongoing studies evaluating the role of CO in gastrointestinal physiology and pathophysiology. RESULTS Carbon monoxide derived from haem oxygenase (HO)-2 is predominantly involved in neuromodulation and in setting the smooth muscle membrane potential, while CO derived from HO-1 has anti-inflammatory and antioxidative properties, which protect gastrointestinal smooth muscle from damage caused by injury or inflammation. Exogenous CO is being explored as a therapeutic agent in a variety of gastrointestinal disorders, including diabetic gastroparesis, post-operative ileus, organ transplantation, inflammatory bowel disease and sepsis. However, identifying the appropriate mechanism for safely delivering CO in humans is a major challenge. CONCLUSIONS Carbon monoxide is an important regulator of gastrointestinal function and protects the gastrointestinal tract against noxious injury. CO is a promising therapeutic target in conditions associated with gastrointestinal injury and inflammation. Elucidating the mechanisms by which CO works and developing safe CO delivery mechanisms are necessary to refine therapeutic strategies.
Collapse
Affiliation(s)
- S J Gibbons
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
34
|
Biliverdin protects against liver ischemia reperfusion injury in swine. PLoS One 2013; 8:e69972. [PMID: 23922878 PMCID: PMC3726748 DOI: 10.1371/journal.pone.0069972] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 06/12/2013] [Indexed: 01/21/2023] Open
Abstract
Ischemia reperfusion injury (IRI) in organ transplantation remains a serious and unsolved problem. Organs that undergo significant damage during IRI, function less well immediately after reperfusion and tend to have more problems at later times when rejection can occur. Biliverdin has emerged as an agent that potently suppress IRI in rodent models. Since the use of biliverdin is being developed as a potential therapeutic modality for humans, we tested the efficacy for its effects on IRI of the liver in swine, an accepted and relevant pre-clinical animal model. Administration of biliverdin resulted in rapid appearance of bilirubin in the serum and significantly suppressed IRI-induced liver dysfunction as measured by multiple parameters including urea and ammonia clearance, neutrophil infiltration and tissue histopathology including hepatocyte cell death. Taken together, our findings, in a large animal model, provide strong support for the continued evaluation of biliverdin as a potential therapeutic in the clinical setting of transplantation of the liver and perhaps other organs.
Collapse
|
35
|
Ryter SW, Choi AMK. Carbon monoxide: present and future indications for a medical gas. Korean J Intern Med 2013; 28:123-40. [PMID: 23525151 PMCID: PMC3604600 DOI: 10.3904/kjim.2013.28.2.123] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/06/2013] [Indexed: 12/30/2022] Open
Abstract
Gaseous molecules continue to hold new promise in molecular medicine as experimental and clinical therapeutics. The low molecular weight gas carbon monoxide (CO), and similar gaseous molecules (e.g., H2S, nitric oxide) have been implicated as potential inhalation therapies in inflammatory diseases. At high concentration, CO represents a toxic inhalation hazard, and is a common component of air pollution. CO is also produced endogenously as a product of heme degradation catalyzed by heme oxygenase enzymes. CO binds avidly to hemoglobin, causing hypoxemia and decreased oxygen delivery to tissues at high concentrations. At physiological concentrations, CO may have endogenous roles as a signal transduction molecule in the regulation of neural and vascular function and cellular homeostasis. CO has been demonstrated to act as an effective anti-inflammatory agent in preclinical animal models of inflammation, acute lung injury, sepsis, ischemia/reperfusion injury, and organ transplantation. Additional experimental indications for this gas include pulmonary fibrosis, pulmonary hypertension, metabolic diseases, and preeclampsia. The development of chemical CO releasing compounds constitutes a novel pharmaceutical approach to CO delivery with demonstrated effectiveness in sepsis models. Current and pending clinical evaluation will determine the usefulness of this gas as a therapeutic in human disease.
Collapse
Affiliation(s)
- Stefan W Ryter
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
36
|
Abstract
Carbon monoxide (CO), a low molecular weight gas, is a ubiquitous environmental product of organic combustion, which is also produced endogenously in the body, as the byproduct of heme metabolism. CO binds to hemoglobin, resulting in decreased oxygen delivery to bodily tissues at toxicological concentrations. At physiological concentrations, CO may have endogenous roles as a potential signaling mediator in vascular function and cellular homeostasis. Exhaled CO (eCO), similar to exhaled nitric oxide (eNO), has been evaluated as a candidate breath biomarker of pathophysiological states, including smoking status, and inflammatory diseases of the lung and other organs. eCO values have been evaluated as potential indicators of inflammation in asthma, stable COPD and exacerbations, cystic fibrosis, lung cancer, or during surgery or critical care. The utility of eCO as a marker of inflammation and its potential diagnostic value remain incompletely characterized. Among other candidate 'medicinal gases' with therapeutic potential, (e.g., NO and H2S), CO has been shown to act as an effective anti-inflammatory agent in preclinical animal models of inflammatory disease, acute lung injury, sepsis, ischemia/reperfusion injury and organ graft rejection. Current and future clinical trials will evaluate the clinical applicability of this gas as a biomarker and/or therapeutic in human disease.
Collapse
Affiliation(s)
- Stefan W Ryter
- Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | | |
Collapse
|
37
|
Noda K, Shigemura N, Tanaka Y, Kawamura T, Hyun Lim S, Kokubo K, Billiar TR, Bermudez CA, Kobayashi H, Nakao A. A novel method of preserving cardiac grafts using a hydrogen-rich water bath. J Heart Lung Transplant 2013; 32:241-50. [DOI: 10.1016/j.healun.2012.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 10/23/2012] [Accepted: 11/04/2012] [Indexed: 02/05/2023] Open
|
38
|
Amano MT, Camara NOS. The immunomodulatory role of carbon monoxide during transplantation. Med Gas Res 2013; 3:1. [PMID: 23295066 PMCID: PMC3582539 DOI: 10.1186/2045-9912-3-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/31/2012] [Indexed: 01/03/2023] Open
Abstract
The number of organ and tissue transplants has increased worldwide in recent decades. However, graft rejection, infections due to the use of immunosuppressive drugs and a shortage of graft donors remain major concerns. Carbon monoxide (CO) had long been regarded solely as a poisonous gas. Ultimately, physiological studies unveiled the endogenous production of CO, particularly by the heme oxygenase (HO)-1 enzyme, recognizing CO as a beneficial gas when used at therapeutic doses. The protective properties of CO led researchers to develop uses for it, resulting in devices and molecules that can deliver CO in vitro and in vivo. The resulting interest in clinical investigations was immediate. Studies regarding the CO/HO-1 modulation of immune responses and their effects on various immune disorders gave rise to transplantation research, where CO was shown to be essential in the protection against organ rejection in animal models. This review provides a perspective of how CO modulates the immune system to improve transplantation and suggests its use as a therapy in the field.
Collapse
Affiliation(s)
- Mariane Tami Amano
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
39
|
Lobb I, Mok A, Lan Z, Liu W, Garcia B, Sener A. Supplemental hydrogen sulphide protects transplant kidney function and prolongs recipient survival after prolonged cold ischaemia-reperfusion injury by mitigating renal graft apoptosis and inflammation. BJU Int 2012; 110:E1187-95. [DOI: 10.1111/j.1464-410x.2012.11526.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
40
|
Schoenfeld MP, Ansari RR, Nakao A, Wink D. A hypothesis on biological protection from space radiation through the use of new therapeutic gases as medical counter measures. Med Gas Res 2012; 2:8. [PMID: 22475015 PMCID: PMC3348081 DOI: 10.1186/2045-9912-2-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 04/04/2012] [Indexed: 12/26/2022] Open
Abstract
Radiation exposure to astronauts could be a significant obstacle for long duration manned space exploration because of current uncertainties regarding the extent of biological effects. Furthermore, concepts for protective shielding also pose a technically challenging issue due to the nature of cosmic radiation and current mass and power constraints with modern exploration technology. The concern regarding exposure to cosmic radiation is biological damage that is associated with increased oxidative stress. It is therefore important and would be enabling to mitigate and/or prevent oxidative stress prior to the development of clinical symptoms and disease. This paper hypothesizes a "systems biology" approach in which a combination of chemical and biological mitigation techniques are used conjunctively. It proposes using new, therapeutic, medical gases as chemical radioprotectors for radical scavenging and as biological signaling molecules for management of the body's response to exposure. From reviewing radiochemistry of water, biological effects of CO, H2, NO, and H2S gas, and mechanisms of radiation biology, it can be concluded that this approach may have therapeutic potential for radiation exposure. Furthermore, it also appears to have similar potential for curtailing the pathogenesis of other diseases in which oxidative stress has been implicated including cardiovascular disease, cancer, chronic inflammatory disease, hypertension, ischemia/reperfusion (IR) injury, acute respiratory distress syndrome, Parkinson's and Alzheimer's disease, cataracts, and aging. We envision applying these therapies through inhalation of gas mixtures or ingestion of water with dissolved gases.
Collapse
Affiliation(s)
- Michael P Schoenfeld
- National Aeronautics and Space Administration Marshall Space Flight Center, Huntsville, Alabama, USA.
| | | | | | | |
Collapse
|
41
|
Hydrogen-enriched preservation protects the isogeneic intestinal graft and amends recipient gastric function during transplantation. Transplantation 2011; 92:985-92. [PMID: 21956195 DOI: 10.1097/tp.0b013e318230159d] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Inhaled hydrogen gas exerts antioxidant and anti-inflammatory effects in rat intestinal transplantation. Here, we investigated whether ex vivo donor organ treatment with dissolved hydrogen would prevent intestinal graft injury. METHODS Isogeneic intestinal transplantation was performed in Lewis rats with vascular flush, luminal preservation, and cold graft storage in nitrogen-bubbled (SITxN2) or hydrogen-bubbled (SITxH2) preservation solution. Lactated Ringer's solution and 3-hr cold ischemia time were used for mechanistic investigations, whereas survival experiments were performed with University of Wisconsin solution and 6-hr cold ischemia time. RESULTS During the early phase of ischemia-reperfusion injury, hydrogen-enriched solution significantly preserved mucosal graft morphology, diminished graft malondialdehyde levels demonstrating substantial reduction potential and blunted proinflammatory molecular responses (early growth response gene [EGR-1], interleukin [IL]-6, IL-1ß, and inducible nitric oxide synthase) within the reperfused intestinal graft muscularis. During the late phase of ischemia-reperfusion injury, circulating IL-6 protein and lactate dehydrogenase levels were significantly ameliorated in SITxH2 animals, which were associated with a favorable functional outcome in in vivo liquid gastrointestinal transit and recipient solid gastric emptying of chrome steel balls, and marked prevention of the posttransplant associated suppression of in vitro muscarinic jejunal contractility. Reflecting improved graft preservation, hydrogen preloading of grafts increased recipient survival rates from 41% to 80%. Anti-inflammatory and antiapoptotic heme oxygenase-1 was significantly upregulated in the hydrogen-treated graft muscularis but not mucosa before reperfusion. CONCLUSIONS Graft preloading with hydrogen demonstrated superior morphologic and functional graft protection in rodent intestinal transplantation, ultimately facilitating recipient survival. Antioxidant capacity and muscularis heme oxygenase-1 upregulation are possible protective mechanisms.
Collapse
|
42
|
Ozaki KS, Yoshida J, Ueki S, Pettigrew GL, Ghonem N, Sico RM, Lee LY, Shapiro R, Lakkis FG, Pacheco-Silva A, Murase N. Carbon monoxide inhibits apoptosis during cold storage and protects kidney grafts donated after cardiac death. Transpl Int 2011; 25:107-17. [PMID: 21999323 DOI: 10.1111/j.1432-2277.2011.01363.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemia/reperfusion (I/R) injury remains as a serious deleterious factor in kidney transplantation (KTx). We hypothesized that carbon monoxide (CO), an endogenous potent cytoprotective molecule, inhibits hypothermia-induced apoptosis of kidney grafts. Using the rat KTx model mimicking the conditions of donation after cardiac death (DCD) as well as nontransplantable human kidney grafts, this study examined effects of CO in preservation solution in improving the quality of marginal kidney grafts. After cardiac cessation, rat kidneys underwent 40 min warm ischemia (WI) and 24 h cold storage (CS) in control UW or UW containing CO (CO-UW). At the end of CS, kidney grafts in control UW markedly increased mitochondrial porin release into the cytosol and resulted in increased cleaved caspase-3 and PARP expression. In contrast, grafts in CO-UW had significantly reduced mitochondrial breakdown and caspase pathway activation. After KTx, recipient survival significantly improved with CO-UW with less TUNEL(+) cells and reduced mRNA upregulation for proinflammatory mediators (IL-6, TNF-α, iNOS). Furthermore, when nontransplantable human kidney grafts were stored in CO-UW for 24 h, graft PARP expression, TUNEL(+) cells, and proinflammatory mediators were less than those in control UW. CO in UW inhibited hypothermia-induced apoptosis and significantly improved kidney graft function and outcomes of KTx.
Collapse
Affiliation(s)
- Kikumi S Ozaki
- Department of Surgery, Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ozaki KS, Kimura S, Murase N. Use of carbon monoxide in minimizing ischemia/reperfusion injury in transplantation. Transplant Rev (Orlando) 2011; 26:125-39. [PMID: 22000659 DOI: 10.1016/j.trre.2011.01.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 01/20/2011] [Indexed: 01/27/2023]
Abstract
Although carbon monoxide (CO) is known to be toxic because of its ability to interfere with oxygen delivery at high concentrations, mammalian cells endogenously generate CO primarily via the catalysis of heme by heme oxygenases. Recent findings have indicated that heme oxygenases and generation of CO serve as a key mechanism to maintain the integrity of the physiological function of organs and supported the development of a new paradigm that CO, at low concentrations, functions as a signaling molecule in the body and exerts significant cytoprotection. Consequently, exogenously delivered CO has been shown to mediate potent protection in various injury models through its anti-inflammatory, vasodilating, and antiapoptotic functions. Ischemia/reperfusion (I/R) injury associated with organ transplantation is one of the major deleterious factors limiting the success of transplantation. Ischemia/reperfusion injury is a complex cascade of interconnected events involving cell damage, apoptosis, vigorous inflammatory responses, microcirculation disturbance, and thrombogenesis. Carbon monoxide has a great potential in minimizing I/R injury. This review will provide an overview of the basic physiology of CO, preclinical studies examining efficacy of CO in I/R injury models, and possible protective mechanisms. Carbon monoxide could be developed to be a valuable therapeutic molecule in minimizing I/R injury in transplantation.
Collapse
Affiliation(s)
- Kikumi S Ozaki
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
44
|
Nakao A, Kaczorowski DJ, Sugimoto R, Billiar TR, McCurry KR. Application of heme oxygenase-1, carbon monoxide and biliverdin for the prevention of intestinal ischemia/reperfusion injury. J Clin Biochem Nutr 2011; 42:78-88. [PMID: 18385824 PMCID: PMC2266059 DOI: 10.3164/jcbn.2008013] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 12/19/2007] [Indexed: 12/31/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury occurs frequently in a variety of clinical settings, including mesenteric artery occlusion, abdominal aneurism surgery, trauma, shock, and small intestinal transplantation, and is associated with substantial morbidity and mortality. Although the exact mechanisms involved in the pathogenesis of intestinal I/R injury have not been fully elucidated, it is generally believed that polymorphonuclear neutrophils, pro-inflammatory cytokines, and mediators generated in the setting of oxidative stress, such as reactive oxygen species (ROS), play important roles. Heme oxygenase (HO) is the rate-limiting enzyme that catalyzes the degradation of heme into equimolar quantities of biliverdin and carbon monoxide (CO), while the central iron is released. An inducible form of HO (HO-1), biliverdin, and CO, have been shown to possess generalized endogenous anti-inflammatory activities and provide protection against intestinal I/R injury. Further, recent observations have demonstrated that exogenous HO-1 expression, as well as exogenously administered CO and biliverdin, have potent cytoprotective effects on intestinal I/R injury as well. Here, we summarize the currently available data regarding the role of the HO system in the prevention intestinal I/R injury.
Collapse
Affiliation(s)
- Atsunori Nakao
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
45
|
Caumartin Y, Stephen J, Deng JP, Lian D, Lan Z, Liu W, Garcia B, Jevnikar AM, Wang H, Cepinskas G, Luke PP. Carbon monoxide-releasing molecules protect against ischemia–reperfusion injury during kidney transplantation. Kidney Int 2011; 79:1080-9. [DOI: 10.1038/ki.2010.542] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
46
|
Varga J, Tóth S, Staško P, Bujdoš M, Veselá J, Jonecová Z, Pomfy M. Different ischemic preconditioning regimens affecting preservation injury of intestines. ACTA ACUST UNITED AC 2011; 46:207-13. [PMID: 21525771 DOI: 10.1159/000327396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 03/08/2011] [Indexed: 11/19/2022]
Abstract
Decreasing ischemia-reperfusion injury in intestinal transplantation is of paramount importance for improving graft recovery and function. This study explores the ability of two ischemic preconditioning (IPC) regimens to reduce preservation injury. Sprague-Dawley rats were divided into 3 groups (n = 11 each). In the controls (group C), intestinal grafts were harvested and preserved. IPC was performed either through 4 cycles of mesenteric ischemia of 4 min each followed by 10 min of reperfusion (group BIPC) or 2 ischemic cycles of 12 min each followed by 10 min of reperfusion (group LIPC). Grafts were stored in histidine-tryptophan-ketoglutarate, and samples were taken 0, 3, 6, 9, 12, 18, and 24 h after preservation. Preservation injury was scored using the Park/Chiu scale. Goblet cells (GC), enteroendocrine cells (EEC) and serotonin-producing EEC (SPEEC) were studied for evaluation of the graft conditions. Group C had the most advanced preservation injury followed by group BIPC. GC count was lowest in group C, followed by BIPC. Comparison between groups BIPC and LIPC showed superior parameters (preservation injury, GC, EEC, and SPEEC) in LIPC. In conclusion, an IPC regimen of 2 ischemic cycles of 12 min each followed by 10 min of reperfusion distinctly decreased the preservation injury of intestinal grafts compared with non-manipulated grafts.
Collapse
Affiliation(s)
- Ján Varga
- Department of Histology and Embryology, Faculty of Medicine, P.J. Šafárik University, Košice, Slovakia.
| | | | | | | | | | | | | |
Collapse
|
47
|
Takagi T, Naito Y, Uchiyama K, Yoshikawa T. The role of heme oxygenase and carbon monoxide in inflammatory bowel disease. Redox Rep 2011; 15:193-201. [PMID: 21062534 DOI: 10.1179/174329210x12650506623889] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease, is a chronic and recurrent inflammatory disorder of the intestinal tract. Since the precise pathogenesis of IBD remains unclear, it is important to investigate the pathogenesis of IBD and to evaluate new anti-inflammatory strategies. Recent evidence suggests that heme oxygenase-1 (HO-1) plays a critical protective role during the development of intestinal inflammation. In fact, it has been demonstrated that the activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in various animal intestinal injury models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid or dextran sulfate sodium. In addition, carbon monoxide (CO) derived from HO-1 has been shown to be involved in the regulation of intestinal inflammation. Furthermore, administration of a low concentration of exogenous CO has a protective effect against intestinal inflammation. These data suggest that HO-1 and CO may be novel therapeutic molecules for patients with gastrointestinal inflammatory diseases. In this review, we present what is currently known regarding the role of HO-1 and CO in intestinal inflammation.
Collapse
Affiliation(s)
- Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | |
Collapse
|
48
|
Blancou P, Tardif V, Simon T, Rémy S, Carreño L, Kalergis A, Anegon I. Immunoregulatory properties of heme oxygenase-1. Methods Mol Biol 2011; 677:247-268. [PMID: 20941616 DOI: 10.1007/978-1-60761-869-0_18] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. A growing body of evidence points toward the capacity of this molecule to inhibit immune reactions and the pivotal role of HO-1 in inflammatory diseases. We will first review the physiological role of HO-1 as determined by the analysis of HO-1-deficient individuals. This will be followed by an examination of the effect of HO-1 within immunopathological contexts such as immune disorders (autoimmunity and allergy) or infections. A section will be devoted to the use of an HO-1 inducer as an immunosuppressive molecule in transplantation. Finally, we will review the molecular basis of HO-1 actions on different immune cells.
Collapse
|
49
|
Nakao A, Billiar TR, Toyoda Y. In response to Schibilsky et al. J Heart Lung Transplant 2010. [DOI: 10.1016/j.healun.2010.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
50
|
Nakao A, Huang CS, Stolz DB, Wang Y, Franks JM, Tochigi N, Billiar TR, Toyoda Y, Tzeng E, McCurry KR. Ex vivo carbon monoxide delivery inhibits intimal hyperplasia in arterialized vein grafts. Cardiovasc Res 2010; 89:457-63. [PMID: 20851811 DOI: 10.1093/cvr/cvq298] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIMS Veins are still the best conduits available for arterial bypass surgery. When these arterialized vein grafts fail, it is often due to the development of intimal hyperplasia (IH). We investigated the feasibility and efficacy of the ex vivo pre-treatment of vein grafts with soluble carbon monoxide (CO) in the inhibition of IH. METHODS AND RESULTS The inferior vena cava was excised from donor rats and placed as an interposition graft into the abdominal aorta of syngeneic rats. Prior to implantation, vein grafts were stored in cold Lactated Ringer (LR) solution with or without CO saturation (bubbling of 100% CO) for 2 h. Three and 6 weeks following grafting, vein grafts treated with cold LR for 2 h developed IH, whereas grafts implanted immediately after harvest demonstrated significantly less IH. Treatment in CO-saturated LR significantly inhibited IH and reduced vascular endothelial cell (VEC) apoptosis. Electron microscopy revealed improved VEC integrity with less platelet/white blood cell aggregation in CO-treated grafts. The effects of CO in preventing IH were associated with activation of hypoxia inducible factor-1α (HIF-1α) and an increase in vascular endothelial growth factor (VEGF) expression at 3-6 h after grafting. Treatment with a HIF-1α inhibitor completely abrogated the induction of VEGF by CO and reversed the protective effects of CO on prevention of IH. CONCLUSION Ex vivo treatment of vein grafts in CO-saturated LR preserved VEC integrity perioperatively and significantly reduced neointima formation. These effects appear to be mediated through the activation of the HIF1α/VEGF pathway.
Collapse
Affiliation(s)
- Atsunori Nakao
- Department of Surgery, University of Pittsburgh Medical Center, E1551, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|