1
|
Gagarinskiy EL, Sharapov MG, Goncharov RG, Gurin AE, Ugraitskaya SV, Fesenko EE. The effectiveness of prolonged hypothermic preservation of isolated rat hearts using oxygen, medical nitrous oxide and carbon monoxide gas mixtures. Arch Biochem Biophys 2025; 765:110295. [PMID: 39798642 DOI: 10.1016/j.abb.2025.110295] [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: 07/08/2024] [Revised: 11/23/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
The possibility of using an oxygen-nitrous oxide mixture for prolonged hypothermic preservation of rat heart for 24 h was investigated. A comparative analysis of restoration of functional activity of hearts in the groups of 24-h preservation at +4 °C with different gases (O2, N2) and gas mixtures (CO + O2, N2O + O2, N2+O2, N2O + N2) was carried out. It was shown that the presence of oxygen in the gas mixture was the key factor for heart preservation. No stable heart preservation was observed in oxygen-free mixtures. At the same time, preservation in pure oxygen showed a significantly lower level of cardiac recovery compared to preservation in gas mixtures O2+CO (6.5 atm.) and O2+N2O (6.5 atm.). LVDP (left ventricular developed pressure) values were 30 ± 19 mmHg and 46 ± 9 mmHg, respectively, with no significant differences found. The decrease in LDVP after 24 h of storage was 26-40 % of the intact control. The results obtained indicate the presence of pronounced synergistic effects of both gases during 24-h heart preservation, which is confirmed by data of marker genes Nfe2l2, Nox1, Prdx1, Hif1a, Nos2, Slc2a4, Ucp-1, Jun, Casp3 expression analysis and myocardial infarction damage level data. The more frequent occurrence of arrhythmias was observed in the oxygen-nitrous oxide group compared with the CO group, and the mechanism of this phenomenon is unclear. Nevertheless, the already medically approved N2O + O2 gas mixture could serve as a balanced choice for future improvements, offering a shorter duration of cardiac preservation compared to the CO + O2 mixture, while ensuring safety in its use.
Collapse
Affiliation(s)
- Evgeniy L Gagarinskiy
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Mars G Sharapov
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Ruslan G Goncharov
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Artem E Gurin
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Svetlana V Ugraitskaya
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Eugeny E Fesenko
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| |
Collapse
|
2
|
Dugbartey GJ. Nitric oxide in kidney transplantation. Biomed Pharmacother 2023; 167:115530. [PMID: 37722191 DOI: 10.1016/j.biopha.2023.115530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023] Open
Abstract
Kidney transplantation is the treatment of choice for patients with kidney failure. Compared to dialysis therapy, it provides better quality of life and confers significant survival advantage at a relatively lower cost. However, the long-term success of this life-saving intervention is severely hampered by an inexorable clinical problem referred to as ischemia-reperfusion injury (IRI), and increases the incidence of post-transplant complications including loss of renal graft function and death of transplant recipients. Burgeoning evidence shows that nitric oxide (NO), a poisonous gas at high concentrations, and with a historic negative public image as an environmental pollutant, has emerged as a potential candidate that holds clinical promise in mitigating IRI and preventing acute and chronic graft rejection when it is added to kidney preservation solutions at low concentrations or when administered to the kidney donor prior to kidney procurement and to the recipient or to the reperfusion circuit at the start and during reperfusion after renal graft preservation. Interestingly, dysregulated or abnormal endogenous production and metabolism of NO is associated with IRI in kidney transplantation. From experimental and clinical perspectives, this review presents endogenous enzymatic production of NO as well as its exogenous sources, and then discusses protective effects of constitutive nitric oxide synthase (NOS)-derived NO against IRI in kidney transplantation via several signaling pathways. The review also highlights a few isolated studies of renal graft protection by NO produced by inducible NOS.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
3
|
Renoprotective effect of Tanshinone IIA against kidney injury induced by ischemia-reperfusion in obese rats. Aging (Albany NY) 2022; 14:8302-8320. [DOI: 10.18632/aging.204304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
|
4
|
Kwong AM, Luke PPW, Bhattacharjee RN. Carbon monoxide mechanism of protection against renal ischemia and reperfusion injury. Biochem Pharmacol 2022; 202:115156. [PMID: 35777450 DOI: 10.1016/j.bcp.2022.115156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022]
Abstract
Carbon monoxide is quickly moving past its historic label as a molecule once feared, to a therapeutic drug that modulates inflammation. The development of carbon monoxide releasing molecules and utilization of heme oxygenase-1 inducers have shown carbon monoxide to be a promising therapy in reducing renal ischemia and reperfusion injury and other inflammatory diseases. In this review, we will discuss the developments and application of carbon monoxide releasing molecules in renal ischemia and reperfusion injury, and transplantation. We will review the anti-inflammatory mechanisms of carbon monoxide in respect to mitigating apoptosis, suppressing dendritic cell maturation and signalling, inhibiting toll-like receptor activation, promoting anti-inflammatory responses, and the effects on renal vasculature.
Collapse
Affiliation(s)
- Aaron M Kwong
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Patrick P W Luke
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Surgery, London Health Sciences Centre, Canada; Matthew Mailing Centre for Translational Transplantation Studies, Canada.
| | - Rabindra N Bhattacharjee
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Surgery, London Health Sciences Centre, Canada; Matthew Mailing Centre for Translational Transplantation Studies, Canada.
| |
Collapse
|
5
|
Tai H, Tong YJ, Yu R, Yu Y, Yao SC, Li LB, Liu Y, Cui XZ, Kuang JS, Meng XS, Jiang XL. A possible new activator of PI3K-Huayu Qutan Recipe alleviates mitochondrial apoptosis in obesity rats with acute myocardial infarction. J Cell Mol Med 2022; 26:3423-3445. [PMID: 35567290 PMCID: PMC9189350 DOI: 10.1111/jcmm.17353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 01/20/2023] Open
Abstract
Obesity, which has unknown pathogenesis, can increase the frequency and seriousness of acute myocardial infarction (AMI). This study evaluated effect of Huayu Qutan Recipe (HQR) pretreatment on myocardial apoptosis induced by AMI by regulating mitochondrial function via PI3K/Akt/Bad pathway in rats with obesity. For in vivo experiments, 60 male rats were randomly divided into 6 groups: sham group, AMI group, AMI (obese) group, 4.5, 9.0 and 18.0 g/kg/d HQR groups. The models fed on HQR with different concentrations for 2 weeks before AMI. For in vitro experiments, the cardiomyocytes line (H9c2) was used. Cells were pretreated with palmitic acid (PA) for 24 h, then to build hypoxia model followed by HQR‐containing serum for 24 h. Related indicators were also detected. In vivo, HQR can lessen pathohistological damage and apoptosis after AMI. In addition, HQR improves blood fat levels, cardiac function, inflammatory factor, the balance of oxidation and antioxidation, as well as lessen infarction in rats with obesity after AMI. Meanwhile, HQR can diminish myocardial cell death by improving mitochondrial function via PI3K/Akt/Bad pathway activation. In vitro, HQR inhibited H9c2 cells apoptosis, improved mitochondrial function and activated the PI3K/Akt/Bad pathway, but effects can be peripeteiad by LY294002. Myocardial mitochondrial dysfunction occurs following AMI and can lead to myocardial apoptosis, which can be aggravated by obesity. HQR can relieve myocardial apoptosis by improving mitochondrial function via the PI3K/Akt/Bad pathway in rats with obesity.
Collapse
Affiliation(s)
- He Tai
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China.,Department of Internal Medicine, Liaoning Provincial Corps Hospital of Chinese People's Armed Police Forces, Shenyang, China
| | - Yu-Jing Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Yu
- Science and Technology Branch, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - You Yu
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Si-Cheng Yao
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ling-Bing Li
- Department of Graduate School, China PLA General Hospital, Beijing, China
| | - Ye Liu
- Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Zheng Cui
- Cardiovascular Surgery, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Jin-Song Kuang
- Department of Endocrinology and Metabolism, The Fourth People's Hospital of Shenyang, Shenyang, China
| | - Xian-Sheng Meng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xiao-Lin Jiang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Nephrology Laboratory, The fourth of Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
6
|
Mansour AM, Khaled RM, Khaled E, Ahmed SK, Ismael OS, Zeinhom A, Magdy H, Ibrahim SS, Abdelfatah M. Ruthenium(II) carbon monoxide releasing molecules: Structural perspective, antimicrobial and anti-inflammatory properties. Biochem Pharmacol 2022; 199:114991. [DOI: 10.1016/j.bcp.2022.114991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 01/12/2023]
|
7
|
Liu Q, Cheng A, Wang Y, Lv Y, Chen Z. Carbon Monoxide in Renal Physiology, Pathogenesis and Treatment of Renal Disease. Curr Pharm Des 2021; 27:4253-4260. [PMID: 34779366 DOI: 10.2174/1381612827666210706161207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/10/2021] [Indexed: 11/22/2022]
Abstract
Carbon monoxide (CO) is one of the endogenous gaseous messengers or gasotransmitters, and is a paramount mediator in physiological and disease conditions. In this review, we focus on the functions of CO in normal and pathological renal physiology. We discuss endogenous renal CO production and signaling in the normal kidney, the characteristic of CO-releasing molecules (CORMs) modalities, and outline its regulatory functions in renal physiology. This article summarizes the mechanisms as well as the effect of CO in the evolving field of renal diseases. We predict numerous innovative CO applications forevolvingcutting-edge scholarly work in the future.
Collapse
Affiliation(s)
- Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anying Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiru Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongman Lv
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
8
|
Dugbartey GJ. Carbon Monoxide in Pancreatic Islet Transplantation: A New Therapeutic Alternative to Patients With Severe Type 1 Diabetes Mellitus. Front Pharmacol 2021; 12:750816. [PMID: 34707503 PMCID: PMC8542862 DOI: 10.3389/fphar.2021.750816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022] Open
Abstract
Pancreatic islet transplantation is a minimally invasive procedure to replace β-cells in a subset of patients with autoimmune type 1 diabetic mellitus, who are extremely sensitive to insulin and lack counter-regulatory measures, and thereby increasing their risk of neuroglycopenia and hypoglycemia unawareness. Thus, pancreatic islet transplantation restores normoglycemia and insulin independence, and prevents long-term surgical complications associated with whole-organ pancreas transplantation. Nonetheless, relative inefficiency of islet isolation and storage process as well as progressive loss of islet function after transplantation due to unvoidable islet inflammation and apoptosis, hinder a successful islet transplantation. Carbon monoxide (CO), a gas which was once feared for its toxicity and death at high concentrations, has recently emerged as a medical gas that seems to overcome the challenges in islet transplantation. This minireview discusses recent findings about CO in preclinical pancreatic islet transplantation and the underlying molecular mechanisms that ensure islet protection during isolation, islet culture, transplantation and post-transplant periods in type 1 diabetic transplant recipients. In addition, the review also discusses clinical translation of these promising experimental findings that serve to lay the foundation for CO in islet transplantation to replace the role of insulin therapy, and thus acting as a cure for type 1 diabetes mellitus and preventing long-term diabetic complications.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON, Canada.,Multi-Organ Transplant Program, London Health Sciences Center, Western University, London, ON, Canada.,Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| |
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
|
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
|
11
|
CO-Releasing Molecule-2 Prevents Acute Kidney Injury through Suppression of ROS-Fyn-ER Stress Signaling in Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9947772. [PMID: 34326922 PMCID: PMC8277502 DOI: 10.1155/2021/9947772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) most commonly appears in critically ill patients in hospitals. AKI is characterized as a quick deterioration of kidney function and has recently been identified to be tightly interlinked with chronic kidney diseases. The emerging major mediators of AKI include oxidative stress and endoplasmic reticulum (ER) stress. Carbon monoxide (CO) attenuates oxidative stress and ER stress in various cells, while Fyn, a member of the Src kinase family, is activated by oxidative stress and contributes to ER stress in skeletal muscle. Considering these, the objective of the current research was to determine (i) the involvement of Fyn in ER stress-mediated AKI and (ii) the effect of CO-releasing molecule-2 (CORM2) on reactive oxygen species- (ROS-) Fyn-ER stress-mediated AKI. Pretreatment with CORM2 (30 mg/kg) efficiently inhibited LPS (30 mg/kg)-induced oxidative stress, inflammation, and cellular apoptosis during AKI in C57BL/6J mice. Also, CORM2 efficiently suppressed the activation of Fyn and ER stress in AKI mice. Consistently, pretreatment with CORM2 inhibited oxidative stress, Fyn activation, ER stress, inflammation, and apoptosis in LPS- or H2O2-stimulated proximal epithelial tubular cells. Fyn inhibition using siRNA or an inhibitor (PP2) significantly attenuated ER stress responses in the cells. These data suggest that CORM2 may become a potential treatment option against ROS-Fyn-ER stress-mediated AKI.
Collapse
|
12
|
Tai H, Jiang XL, Song N, Xiao HH, Li Y, Cheng MJ, Yin XM, Chen YR, Yang GL, Jiang XY, Kuang JS, Lan ZM, Jia LQ. Tanshinone IIA Combined With Cyclosporine A Alleviates Lung Apoptosis Induced by Renal Ischemia-Reperfusion in Obese Rats. Front Med (Lausanne) 2021; 8:617393. [PMID: 34012969 PMCID: PMC8126627 DOI: 10.3389/fmed.2021.617393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/19/2021] [Indexed: 11/16/2022] Open
Abstract
Acute lung injury (ALI), which is induced by renal ischemia-reperfusion (IR), is one of the leading causes of acute renal IR-related death. Obesity raises the frequency and severity of acute kidney injury (AKI) and ALI. Tanshinone IIA (TIIA) combined with cyclosporine A (CsA) was employed to lessen the lung apoptosis led by renal IR and to evaluate whether TIIA combined with CsA could alleviate lung apoptosis by regulating mitochondrial function through the PI3K/Akt/Bad pathway in obese rats. Hematoxylin-eosin (HE) staining was used to assess the histology of the lung injury. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) was used to assess apoptosis of the lung. Electron microscopy was used to assess mitochondrial morphology in lung cells. Arterial blood gas and pulmonary function were used to assess the external respiratory function. Mitochondrial function was used to assess the internal respiratory function and mitochondrial dynamics and biogenesis. Western blot (WB) was used to examine the PI3K/Akt/Bad pathway-related proteins. TIIA combined with CsA can alleviate lung apoptosis by regulating mitochondrial function through the PI3K/Akt/Bad pathway in obese rats.
Collapse
Affiliation(s)
- He Tai
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiao-Lin Jiang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Nephrology, The Fourth of Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Nan Song
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hong-He Xiao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Yue Li
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Mei-Jia Cheng
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiao-Mei Yin
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yi-Ran Chen
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Guan-Lin Yang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiao-Yu Jiang
- Department of Foreign Languages, Dalian Medical University, Dalian, China
| | - Jin-Song Kuang
- Department of Endocrinology and Metabolism, The Fourth People's Hospital of Shenyang, Shenyang, China
| | - Zhi-Ming Lan
- Department of Medical Laboratory, The Fourth Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Lian-Qun Jia
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
13
|
Tai H, Jiang XL, Lan ZM, Li Y, Kong L, Yao SC, Song N, Lv MJ, Wu J, Yang P, Xiao XS, Yang GL, Kuang JS, Jia LQ. Tanshinone IIA combined with CsA inhibit myocardial cell apoptosis induced by renal ischemia-reperfusion injury in obese rats. BMC Complement Med Ther 2021; 21:100. [PMID: 33752661 PMCID: PMC7986523 DOI: 10.1186/s12906-021-03270-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Acute myocardial injury (AMI), which is induced by renal ischemia-reperfusion (IR), is a significant cause of acute kidney injury (AKI)-related associated death. Obesity increases the severity and frequency of AMI and AKI. Tanshinone IIA (TIIA) combined with cyclosporine A (CsA) pretreatment was used to alleviate myocardial cell apoptosis induced by renal IR, and to determine whether TIIA combined with CsA would attenuate myocardial cell apoptosis by modulating mitochondrial function through the PI3K/Akt/Bad pathway in obese rats. METHODS Male rates were fed a high fat diet for 8 weeks to generate obesity. AKI was induced by 30 min of kidney ischemia followed 24 h of reperfusion. Obese rats were given TIIA (10 mg/kg·d) for 2 weeks and CsA (5 mg/kg) 30 min before renal IR. After 24 h of reperfusion, the rats were anaesthetized, the blood were fetched from the abdominal aorta and kidney were fetched from abdominal cavity, then related indicators were examined. RESULTS TIIA combined with CsA can alleviate the pathohistological injury and apoptosis induced by renal IR in myocardial cells. TIIA combined with CsA improved cardiac function after renal ischemia (30 min)-reperfusion (24 h) in obese rats. At the same time, TIIA combined with CsA improved mitochondrial function. Abnormal function of mitochondria was supported by decreases in respiration controlling rate (RCR), intracellular adenosine triphosphate (ATP), oxygen consumption rate, and mitochondrial membrane potential (MMP), and increases in mitochondrial reactive oxygen species (ROS), opening of the mitochondrial permeability transition pore (mPTP), mitochondrial DNA damage, and mitochondrial respiratory chain complex enzymes. The injury of mitochondrial dynamic function was assessed by decrease in dynamin-related protein 1 (Drp1), and increases in mitofusin1/2 (Mfn1/2), and mitochondrial biogenesis injury was assessed by decreases in PPARγ coactivator-1-α (PGC-1), nucleo respiratory factor1 (Nrf1), and transcription factor A of mitochondrial (TFam). CONCLUSION We used isolated mitochondria from rat myocardial tissues to demonstrate that myocardial mitochondrial dysfunction occurred along with renal IR to induce myocardial cell apoptosis; obesity aggravated apoptosis. TIIA combined with CsA attenuated myocardial cell apoptosis by modulating mitochondrial function through the PI3K/Akt/Bad pathway in obese rats.
Collapse
Affiliation(s)
- He Tai
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiao-Lin Jiang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Nephrology, The fourth of Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Zhi-Ming Lan
- Department of Medical laboratory, The fourth of Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Yue Li
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Si-Cheng Yao
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Nan Song
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Mei-Jun Lv
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jin Wu
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ping Yang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Liaoning Traditional Chinese Medicine, Shenyang, China
| | - Xuan-Si Xiao
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Guan-Lin Yang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jin-Song Kuang
- Department of Endocrinology and Metabolic, Shenyang the Fourth Hospital of People, Shenyang, China
| | - Lian-Qun Jia
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.
| |
Collapse
|
14
|
Therapeutic Potential of Heme Oxygenase-1 and Carbon Monoxide in Acute Organ Injury, Critical Illness, and Inflammatory Disorders. Antioxidants (Basel) 2020; 9:antiox9111153. [PMID: 33228260 PMCID: PMC7699570 DOI: 10.3390/antiox9111153] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. HO-1 has been implicated as a cytoprotectant in various models of acute organ injury and disease (i.e., lung, kidney, heart, liver). Thus, HO-1 may serve as a general therapeutic target in inflammatory diseases. HO-1 may function as a pleiotropic modulator of inflammatory signaling, via the removal of heme, and generation of its enzymatic degradation-products. Iron release from HO activity may exert pro-inflammatory effects unless sequestered, whereas BV/BR have well-established antioxidant properties. CO, derived from HO activity, has been identified as an endogenous mediator that can influence mitochondrial function and/or cellular signal transduction programs which culminate in the regulation of apoptosis, cellular proliferation, and inflammation. Much research has focused on the application of low concentration CO, whether administered in gaseous form by inhalation, or via the use of CO-releasing molecules (CORMs), for therapeutic benefit in disease. The development of novel CORMs for their translational potential remains an active area of investigation. Evidence has accumulated for therapeutic effects of both CO and CORMs in diseases associated with critical care, including acute lung injury/acute respiratory distress syndrome (ALI/ARDS), mechanical ventilation-induced lung injury, pneumonias, and sepsis. The therapeutic benefits of CO may extend to other diseases involving aberrant inflammatory processes such as transplant-associated ischemia/reperfusion injury and chronic graft rejection, and metabolic diseases. Current and planned clinical trials explore the therapeutic benefit of CO in ARDS and other lung diseases.
Collapse
|
15
|
Levin N, Marcolongo JP, Cadranel A, Slep LD. Time-Resolved Exploration of a photoCORM {Ru(bpy)} Model Compound. Inorg Chem 2020; 59:12075-12085. [DOI: 10.1021/acs.inorgchem.0c01025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Natalia Levin
- Facultad de Ciencias Exactas y Naturales, Departamento de Quı́mica Inorgánica, Analı́tica y Quı́mica Fı́sica, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Instituto de Quı́mica-Fı́sica de Materiales, Medio Ambiente y Energı́a, CONICET, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Max Planck Institute for Chemical Energy Conversion, Stiftstr. 34−36, 45470 Mülheim an der Ruhr, Germany
| | - Juan Pablo Marcolongo
- Facultad de Ciencias Exactas y Naturales, Departamento de Quı́mica Inorgánica, Analı́tica y Quı́mica Fı́sica, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Instituto de Quı́mica-Fı́sica de Materiales, Medio Ambiente y Energı́a, CONICET, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Alejandro Cadranel
- Facultad de Ciencias Exactas y Naturales, Departamento de Quı́mica Inorgánica, Analı́tica y Quı́mica Fı́sica, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Instituto de Quı́mica-Fı́sica de Materiales, Medio Ambiente y Energı́a, CONICET, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Department of Chemistry and Pharmacy & Interdisciplinary Center for Molecular Materials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Egerlandstrasse 3, 91058 Erlangen, Germany
| | - Leonardo Daniel Slep
- Facultad de Ciencias Exactas y Naturales, Departamento de Quı́mica Inorgánica, Analı́tica y Quı́mica Fı́sica, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
- Instituto de Quı́mica-Fı́sica de Materiales, Medio Ambiente y Energı́a, CONICET, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| |
Collapse
|
16
|
Fesenko EE, Gagarinsky EL, Averin AS, Grudinin NV, Gurin AE, Shishova NV, Shvirst NE, Goltyaev MV, Kovtun AL. The Condition of the Rat Myocardium and Isolated Sheep Heart after Prolonged 24-Hour Hypothermic Preservation in a Pressurized Carbon Monoxide–Oxygen Gas Mixture. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920040065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
17
|
Ostróżka-Cieślik A, Dolińska B. The Role of Hormones and Trophic Factors as Components of Preservation Solutions in Protection of Renal Function before Transplantation: A Review of the Literature. Molecules 2020; 25:E2185. [PMID: 32392782 PMCID: PMC7248710 DOI: 10.3390/molecules25092185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Transplantation is currently a routine method for treating end-stage organ failure. In recent years, there has been some progress in the development of an optimal composition of organ preservation solutions, improving the vital functions of the organ and allowing to extend its storage period until implantation into the recipient. Optimizations are mostly based on commercial solutions, routinely used to store grafts intended for transplantation. The paper reviews hormones with a potential nephroprotective effect, which were used to modify the composition of renal perfusion and preservation solutions. Their effectiveness as ingredients of preservation solutions was analysed based on a literature review. Hormones and trophic factors are innovative preservation solution supplements. They have a pleiotropic effect and affect normal renal function. The expression of receptors for melatonin, prolactin, thyrotropin, corticotropin, prostaglandin E1 and trophic factors was confirmed in the kidneys, which suggests that they are a promising therapeutic target for renal IR (ischemia-reperfusion) injury. They can have anti-inflammatory, antioxidant and anti-apoptotic effects, limiting IR injury.
Collapse
Affiliation(s)
- Aneta Ostróżka-Cieślik
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| | - Barbara Dolińska
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland
| |
Collapse
|
18
|
CORM-2-Solid Lipid Nanoparticles Maintain Integrity of Blood-Spinal Cord Barrier After Spinal Cord Injury in Rats. Mol Neurobiol 2020; 57:2671-2689. [DOI: 10.1007/s12035-020-01914-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 03/30/2020] [Indexed: 12/21/2022]
|
19
|
Canesin G, Hejazi SM, Swanson KD, Wegiel B. Heme-Derived Metabolic Signals Dictate Immune Responses. Front Immunol 2020; 11:66. [PMID: 32082323 PMCID: PMC7005208 DOI: 10.3389/fimmu.2020.00066] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022] Open
Abstract
Heme is one of the most abundant molecules in the body acting as the functional core of hemoglobin/myoglobin involved in the O2/CO2 carrying in the blood and tissues, redox enzymes and cytochromes in mitochondria. However, free heme is toxic and therefore its removal is a significant priority for the host. Heme is a well-established danger-associated molecular pattern (DAMP), which binds to toll-like receptor 4 (TLR4) to induce immune responses. Heme-derived metabolites including the bile pigments, biliverdin (BV) and bilirubin (BR), were first identified as toxic drivers of neonatal jaundice in 1800 but have only recently been appreciated as endogenous drivers of multiple signaling pathways involved in protection from oxidative stress and regulators of immune responses. The tissue concentration of heme, BV and BR is tightly controlled. Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. BLVR-A is a fascinating protein that possesses a classical protein kinase domain, which is activated in response to BV binding to its enzymatic site and initiates the downstream mitogen-activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways. This links BLVR-A activity to cell growth and survival pathways. BLVR-A also contains a bZip DNA binding domain and a nuclear export sequence (NES) and acts as a transcription factor to regulate the expression of immune modulatory genes. Here we will discuss the role of heme-related immune response and the potential for targeting the heme system for therapies directed toward hepatitis and cancer.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Seyed M. Hejazi
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Kenneth D. Swanson
- Brain Tumor Center and Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Barbara Wegiel
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Goebel U, Wollborn J. Carbon monoxide in intensive care medicine-time to start the therapeutic application?! Intensive Care Med Exp 2020; 8:2. [PMID: 31919605 PMCID: PMC6952485 DOI: 10.1186/s40635-020-0292-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/05/2020] [Indexed: 12/18/2022] Open
Abstract
Carbon monoxide (CO) is not only known as a toxic gas due to its characteristics as an odorless molecule and its rapid binding to haem-containing molecules, thus inhibiting the respiratory chain in cells resulting in hypoxia. For decades, scientists established evidence about its endogenously production in the breakdown of haem via haem-oxygenase (HO-1) and its physiological effects. Among these, the modulation of various systems inside the body are well described (e.g., anti-inflammatory, anti-oxidative, anti-apoptotic, and anti-proliferative). Carbon monoxide is able to modulate several extra- and intra-cellular signaling molecules leading to differentiated response according to the specific stimulus. With our growing understanding in the way CO exerts its effects, especially in the mitochondria and its intracellular pathways, it is tempting to speculate about a clinical application of this substance. Since HO-1 is not easy to induce, research focused on the application of the gaseous molecule CO by itself or the implementation of carbon monoxide releasing molecules (CO-RM) to deliver the molecule at a time- and dose dependently safe way to any target organ. After years of research in cellular systems and animal models, summing up data about safety issues as well as possible target to treat in various diseases, the first feasibility trials in humans were established. Up-to-date, safety issues have been cleared for low-dose carbon monoxide inhalation (up to 500 ppm), while there is no clinical data regarding the injection or intake of any kind of CO-RM so far. Current models of human research include sepsis, acute lung injury, and acute respiratory distress syndrome as well as acute kidney injury. Carbon monoxide is a most promising candidate in terms of a therapeutic agent to improve outbalanced organ conditions. In this paper, we summarized the current understanding of carbon monoxide’s biology and its possible organ targets to treating the critically ill patients in tomorrow’s ICU.
Collapse
Affiliation(s)
- Ulrich Goebel
- Department of Anaesthesiology and Critical Care, St. Franziskus-Hospital, Hohenzollernring 70, 48145, Münster, Germany.
| | - Jakob Wollborn
- Department of Anaesthesiology and Critical Care, Medical Centre - University of Freiburg, Faculty of Medicine, Freiburg im Breisgau, Germany
| |
Collapse
|
21
|
Yang X, de Caestecker M, Otterbein LE, Wang B. Carbon monoxide: An emerging therapy for acute kidney injury. Med Res Rev 2019; 40:1147-1177. [PMID: 31820474 DOI: 10.1002/med.21650] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/31/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
Treating acute kidney injury (AKI) represents an important unmet medical need both in terms of the seriousness of this medical problem and the number of patients. There is also a large untapped market opportunity in treating AKI. Over the years, there has been much effort in search of therapeutics with minimal success. However, over the same time period, new understanding of the underlying pathobiology and molecular mechanisms of kidney injury have undoubtedly helped the search for new therapeutics. Along this line, carbon monoxide (CO) has emerged as a promising therapeutic agent because of its demonstrated cytoprotective, and immunomodulatory effects. CO has also been shown to sensitize cancer, but not normal cells, to chemotherapy. This is particularly important in treating cisplatin-induced AKI, a common clinical problem that develops in patients receiving cisplatin therapies for a number of different solid organ malignancies. This review will examine and make the case that CO be developed into a therapeutic agent against AKI.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Mark de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| |
Collapse
|
22
|
Abstract
BACKGROUND The potential of a mesenchymal stem cell (MSC) therapy to accelerate the repair of ischemically damaged human kidneys during 24 hours of warm perfusion was evaluated. The hypothesis was that by administering MSC directly to the renal tissue, there would be an improved opportunity for cellular repair mediated by intrarenal paracrine effects. METHODS Studies were performed using the exsanguinous metabolic support (EMS) tissue-engineering platform. Five pairs of human kidney allografts from donation after circulatory death donors were studied. One human kidney was EMS perfused for 24 hours (control), whereas its paired kidney was EMS perfused with MSC (1 × 10). The kidneys were evaluated for DNA synthesis, cytokine/chemokine synthesis, cytoskeletal regeneration, and mitosis. RESULTS Treatment with MSC resulted in reduced inflammatory cytokines synthesized by the kidneys. Mesenchymal stem cell treatment led to a significant increase in the synthesis of adenosine triphosphate and growth factors resulting in normalization of metabolism and the cytoskeleton. Toluidine Blue staining of MSC-treated kidneys demonstrated a significant increase in the number of renal cells undergoing mitosis (26%) compared with EMS perfusion alone. CONCLUSIONS To our knowledge, our work is the first to have demonstrated actual renal regeneration while ischemically damaged human kidneys are perfused ex vivo for 24 hours. The observed regeneration entails: increased synthesis of adenosine triphosphate, a reduced inflammatory response, increased synthesis of growth factors, normalization of the cytoskeleton and mitosis. The ability to regenerate renal tissue ex vivo sufficiently to result in immediate function could revolutionize transplantation by solving the chronic organ shortage.
Collapse
|
23
|
Exenatide Reduces Graft Injury in a Rat Transplantation Model Using Kidneys Donated after Cardiac Death. Transplant Proc 2019; 51:2116-2123. [PMID: 31303407 DOI: 10.1016/j.transproceed.2019.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/04/2019] [Accepted: 04/22/2019] [Indexed: 11/21/2022]
Abstract
Besides being used in the therapy of type 2 diabetes, exenatide reduces cerebral ischemia-reperfusion (I/R) injury. We evaluated the potential effects of exenatide on inhibition of apoptosis in kidney grafts donated after cardiac death and on reduction of I/R injury after kidney transplantation (KTx) in a rat model. We used a rat syngeneic KTx model with kidney grafts obtained after cardiac death, and apoptosis was detected in the graft before KTx. Graft function, rat survival, morphologic examination, and activation of inflammatory molecules were analyzed after KTx. By the end of the cold storage, exenatide pretreatment donors had significantly reduced caspase pathway activation, terminal deoxynucleotidyl transferase dUTP nick-end labeling--positive cells, release of mitochondrial porin proteins into the cytosol, and expression of cleaved caspase-3 and poly (ADP-ribose) polymerase in kidney grafts. Exenatide pretreatment improved renal function survival rate with lower scores of acute tubular necrosis, infiltrating macrophages, and interstitial fibrosis as well as reduced messenger RNA expression of inflammatory mediators (tumor necrosis factor α, interleukin-6, interleukin-1β, and intercellular adhesion molecule-1) after KTx. Our study showed that exenatide reduced I/R injury in kidneys donated after cardiac death in a rat transplantation model and improved recipient survival and graft function.
Collapse
|
24
|
Drummond HA, Mitchell ZL, Abraham NG, Stec DE. Targeting Heme Oxygenase-1 in Cardiovascular and Kidney Disease. Antioxidants (Basel) 2019; 8:antiox8060181. [PMID: 31216709 PMCID: PMC6617021 DOI: 10.3390/antiox8060181] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/13/2019] [Accepted: 06/15/2019] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase (HO) plays an important role in the cardiovascular system. It is involved in many physiological and pathophysiological processes in all organs of the cardiovascular system. From the regulation of blood pressure and blood flow to the adaptive response to end-organ injury, HO plays a critical role in the ability of the cardiovascular system to respond and adapt to changes in homeostasis. There have been great advances in our understanding of the role of HO in the regulation of blood pressure and target organ injury in the last decade. Results from these studies demonstrate that targeting of the HO system could provide novel therapeutic opportunities for the treatment of several cardiovascular and renal diseases. The goal of this review is to highlight the important role of HO in the regulation of cardiovascular and renal function and protection from disease and to highlight areas in which targeting of the HO system needs to be translated to help benefit patient populations.
Collapse
Affiliation(s)
- Heather A Drummond
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| | - Zachary L Mitchell
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| | - Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Vahalla, NY 10595, USA.
- Joan C. Edwards School of Medicine, Marshall University, Huntington, VA 25701, USA.
| | - David E Stec
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| |
Collapse
|
25
|
Situmorang GR, Sheerin NS. Ischaemia reperfusion injury: mechanisms of progression to chronic graft dysfunction. Pediatr Nephrol 2019; 34:951-963. [PMID: 29603016 PMCID: PMC6477994 DOI: 10.1007/s00467-018-3940-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/18/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
The increasing use of extended criteria organs to meet the demand for kidney transplantation raises an important question of how the severity of early ischaemic injury influences long-term outcomes. Significant acute ischaemic kidney injury is associated with delayed graft function, increased immune-associated events and, ultimately, earlier deterioration of graft function. A comprehensive understanding of immediate molecular events that ensue post-ischaemia and their potential long-term consequences are key to the discovery of novel therapeutic targets. Acute ischaemic injury primarily affects tubular structure and function. Depending on the severity and persistence of the insult, this may resolve completely, leading to restoration of normal function, or be sustained, resulting in persistent renal impairment and progressive functional loss. Long-term effects of acute renal ischaemia are mediated by several mechanisms including hypoxia, HIF-1 activation, endothelial dysfunction leading to vascular rarefaction, sustained pro-inflammatory stimuli involving innate and adaptive immune responses, failure of tubular cells to recover and epigenetic changes. This review describes the biological relevance and interaction of these mechanisms based on currently available evidence.
Collapse
Affiliation(s)
- Gerhard R Situmorang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Urology Department, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, 10430, Indonesia
| | - Neil S Sheerin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
26
|
Hopper CP, Wollborn J. Delivery of carbon monoxide via halogenated ether anesthetics. Nitric Oxide 2019; 89:93-95. [PMID: 31125687 DOI: 10.1016/j.niox.2019.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Christopher P Hopper
- Department of Medicinal Chemistry, College of Pharmacy, The University of Florida, Gainesville, Florida, USA; Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Germany; Institute for Experimental Biomedicine, University Hospital Wuerzburg, Germany.
| | - Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
27
|
Uddin MJ, Pak ES, Ha H. Carbon monoxide releasing molecule-2 protects mice against acute kidney injury through inhibition of ER stress. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:567-575. [PMID: 30181703 PMCID: PMC6115348 DOI: 10.4196/kjpp.2018.22.5.567] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/28/2018] [Accepted: 07/12/2018] [Indexed: 12/21/2022]
Abstract
Acute kidney injury (AKI), which is defined as a rapid decline of renal function, becomes common and recently recognized to be closely intertwined with chronic kidney diseases. Current treatment for AKI is largely supportive, and endoplasmic reticulum (ER) stress has emerged as a novel mediator of AKI. Since carbon monoxide attenuates ER stress, the objective of the present study aimed to determine the protective effect of carbon monoxide releasing molecule-2 (CORM2) on AKI associated with ER stress. Kidney injury was induced after LPS (15 mg/kg) treatment at 12 to 24 h in C57BL/6J mice. Pretreatment of CORM2 (30 mg/kg) effectively prevented LPS-induced oxidative stress and inflammation during AKI in mice. CORM2 treatment also effectively inhibited LPS-induced ER stress in AKI mice. In order to confirm effect of CO on the pathophysiological role of tubular epithelial cells in AKI, we used mProx24 cells. Pretreatment of CORM2 attenuated LPS-induced ER stress, oxidative stress, and inflammation in mProx24 cells. These data suggest that CO therapy may prevent ER stress-mediated AKI.
Collapse
Affiliation(s)
- Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Eun Seon Pak
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
28
|
Marker SC, MacMillan SN, Zipfel WR, Li Z, Ford PC, Wilson JJ. Photoactivated in Vitro Anticancer Activity of Rhenium(I) Tricarbonyl Complexes Bearing Water-Soluble Phosphines. Inorg Chem 2018; 57:1311-1331. [PMID: 29323880 PMCID: PMC8117114 DOI: 10.1021/acs.inorgchem.7b02747] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fifteen water-soluble rhenium compounds of the general formula [Re(CO)3(NN)(PR3)]+, where NN is a diimine ligand and PR3 is 1,3,5-triaza-7-phosphaadamantane (PTA), tris(hydroxymethyl)phosphine (THP), or 1,4-diacetyl-1,3,7-triaza-5-phosphabicylco[3.3.1]nonane (DAPTA), were synthesized and characterized by multinuclear NMR spectroscopy, IR spectroscopy, and X-ray crystallography. The complexes bearing the THP and DAPTA ligands exhibit triplet-based luminescence in air-equilibrated aqueous solutions with quantum yields ranging from 3.4 to 11.5%. Furthermore, the THP and DAPTA complexes undergo photosubstitution of a CO ligand upon irradiation with 365 nm light with quantum yields ranging from 1.1 to 5.5% and sensitize the formation of 1O2 with quantum yields as high as 70%. In contrast, all of the complexes bearing the PTA ligand are nonemissive and do not undergo photosubstitution upon irradiation with 365 nm light. These compounds were evaluated as photoactivated anticancer agents in human cervical (HeLa), ovarian (A2780), and cisplatin-resistant ovarian (A2780CP70) cancer cell lines. All of the complexes bearing THP and DAPTA exhibited a cytotoxic response upon irradiation with minimal toxicity in the absence of light. Notably, the complex with DAPTA and 1,10-phenanthroline gave rise to an IC50 value of 6 μM in HeLa cells upon irradiation, rendering it the most phototoxic compound in this library. The nature of the photoinduced cytotoxicity of this compound was explored in further detail. These data indicate that the phototoxic response may result from the release of both CO and the rhenium-containing photoproduct, as well as the production of 1O2.
Collapse
Affiliation(s)
- Sierra C. Marker
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Samantha N. MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Warren R. Zipfel
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Zhi Li
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California 93106-9510, United States
| | - Peter C. Ford
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California 93106-9510, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
29
|
Jang HJ, Hong EM, Kim M, Kim JH, Jang J, Park SW, Byun HW, Koh DH, Choi MH, Kae SH, Lee J. Simvastatin induces heme oxygenase-1 via NF-E2-related factor 2 (Nrf2) activation through ERK and PI3K/Akt pathway in colon cancer. Oncotarget 2018; 7:46219-46229. [PMID: 27323826 PMCID: PMC5216792 DOI: 10.18632/oncotarget.10078] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/29/2016] [Indexed: 12/30/2022] Open
Abstract
Statin has been known not only as their cholesterol-lowering action but also on their pleiotropic effects including anti-inflammatory and anti-oxidant as well as anti-cancer effect. Nrf2 (NF-E2-related factor 2) is a transcription factor to activate cellular antioxidant response to oxidative stress. There are little known whether statins affect activation of Nrf2 and Nrf2 signaling pathway in colon cancer cells. We investigated whether simvastatin stimulates the expression of Nrf2 and nuclear translocation of Nrf2 and which signal pathway is involved in the expression of Nrf2 and antioxidant enzymes. We investigated the effect of simvastatin on the expression of Nrf2 and nuclear translocation of Nrf2 in two human colon cancer cell lines, HT-29 and HCT 116 through cell proliferation assay, Western blotting and immunocytochemical analysis. We evaluated which signal pathway such as ERK or PI3K pathway affect Nrf2 activation and whether simvastatin induces antioxidant enzymes (heme oxygenase-1 (HO-1), NAD(P)H: quinine oxidoreductase 1 (NQO1), γ-glutamate-cysteine ligase catalytic subunit (GCLC)). We demonstrated simvastatin-induced dose-dependent up-regulation of Nrf2 expression and stimulated Nrf2 nuclear translocation in colon cancer cells. We also demonstrated that simvastatin-induced anti-oxidant enzymes (HO-1, NQO1, and GCLC) in HT-29 and HCT 116 cells. PI3K/Akt inhibitor (LY294002) and ERK inhibitor (PD98059) suppressed simvastatin-induced Nrf2 and HO-1 expression in both HT-29 and HCT 116 cells. This study shows that simvastatin induces the activation and nuclear translocation of Nrf2 and the expression of various anti-oxidant enzymes via ERK and PI3K/Akt pathway in colon cancer cells.
Collapse
Affiliation(s)
- Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Eun Mi Hong
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Mikang Kim
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Jae Hyun Kim
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Juah Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Hyun Wu Byun
- Division of Gastroenterology, Department of Internal Medicine, Hangang Sacred Heart Hospital, Hallym University School of Medicine, Seoul, Korea
| | - Dong Hee Koh
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Min Ho Choi
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Sea Hyub Kae
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| | - Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Gyeonggi do, Korea
| |
Collapse
|
30
|
Nishida T, Hayashi T, Inamoto T, Kato R, Ibuki N, Takahara K, Takai T, Yoshikawa Y, Uchimoto T, Saito K, Tanda N, Kouno J, Minami K, Uehara H, Hirano H, Nomi H, Okada Y, Azuma H. Dual Gas Treatment With Hydrogen and Carbon Monoxide Attenuates Oxidative Stress and Protects From Renal Ischemia-Reperfusion Injury. Transplant Proc 2018; 50:250-258. [DOI: 10.1016/j.transproceed.2017.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/26/2017] [Accepted: 12/05/2017] [Indexed: 01/14/2023]
|
31
|
Ulbrich F, Hagmann C, Buerkle H, Romao CC, Schallner N, Goebel U, Biermann J. The Carbon monoxide releasing molecule ALF-186 mediates anti-inflammatory and neuroprotective effects via the soluble guanylate cyclase ß1 in rats' retinal ganglion cells after ischemia and reperfusion injury. J Neuroinflammation 2017; 14:130. [PMID: 28655348 PMCID: PMC5488359 DOI: 10.1186/s12974-017-0905-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 06/18/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The endogenously produced gaseous molecule carbon monoxide is able to promote organ protection after ischemia-reperfusion injuries (IRI). The impact of carbon monoxide releasing molecules (CORM) regarding inflammation in neuronal tissues has not been studied in detail. In this investigation, we aimed to analyze the effects of the CORM ALF-186 on neuro-inflammation and hypothesized that the soluble guanylate cyclase (sGC) is playing a decisive role. METHODS Retinal ischemia-reperfusion injury was performed for 60 min in Sprague-Dawley rats. Thereafter, the CORM ALF-186 (10 mg/kg) in the presence or absence of the sGC inhibitor ODQ was injected via a tail vein. Retinal tissue was harvested 24 h later to analyze mRNA or protein expression of sGC-β1 subunit, transcription factors NF-κB and CREB, the inflammatory cytokines TNF-α and IL-6, as well as the heat shock proteins (HSP) HSP-70 and HSP-90. Immunohistochemistry was performed on frozen sections of the retina. The overall neuroprotective effect of ALF-186 was assessed by counting fluorogold-pre-labeled retinal ganglion cells (RGC) 7 days after IRI. RESULTS Ischemia-reperfusion mediated loss of vital RGC was attenuated by the administration of ALF-186 after injury. ALF-186 treatment after IRI induced sGC-ß1 leading to a decreased NF-κB and CREB phosphorylation. Consecutively, ALF-186 mitigated IRI induced TNF-α and IL-6 expression in the retina and in the rats' serum. Moreover, ALF-186 attenuated heat shock protein 70 (Hsp-70) while increasing Hsp-90. The sGC-inhibitor ODQ attenuated the anti-inflammatory effects of ALF-186 and increased retinal loss of ganglion cells. These results were confirmed by immunohistochemistry. CONCLUSION The CORM ALF-186 protected RGC from IRI induced loss. Furthermore, ALF-186 reduced IRI mediated neuroinflammation in the retina and in the serum by activating sGC. Inhibition of sGC stopped the beneficial and protective effects of ALF-186. ALF-186 may present a promising therapeutic alternative in treating inflammation after neuronal IRI.
Collapse
Affiliation(s)
- Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany
| | - Claus Hagmann
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany
| | - Carlos C Romao
- Instituto de Tecnologia Química e Biológica-António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- Alfama Ltd., Instituto de Biologia Experimental e Tecnológica, IBET, Oeiras, Portugal
| | - Nils Schallner
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, D-79106, Freiburg, Germany.
| | - Julia Biermann
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
32
|
Protective effect of zerumbone reduces lipopolysaccharide-induced acute lung injury via antioxidative enzymes and Nrf2/HO-1 pathway. Int Immunopharmacol 2017; 46:194-200. [DOI: 10.1016/j.intimp.2017.03.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/17/2017] [Accepted: 03/07/2017] [Indexed: 01/08/2023]
|
33
|
|
34
|
Üstün E, Özgür A, Coşkun KA, Demir S, Özdemir İ, Tutar Y. CO-releasing properties and anticancer activities of manganese complexes with imidazole/benzimidazole ligands. J COORD CHEM 2016; 69:3384-3394. [DOI: 10.1080/00958972.2016.1231921] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/27/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Elvan Üstün
- Faculty of Art and Science, Department of Chemistry, Ordu University, Ordu, Turkey
| | - Aykut Özgür
- Faculty of Natural Sciences and Engineering, Department of Bioengineering, Gaziosmanpaşa University, Tokat, Turkey
| | - Kübra A. Coşkun
- Faculty of Natural Sciences and Engineering, Department of Bioengineering, Gaziosmanpaşa University, Tokat, Turkey
| | - Serpil Demir
- Faculty of Science, Department of Chemistry, İnönü University, Malatya, Turkey
| | - İsmail Özdemir
- Faculty of Science, Department of Chemistry, İnönü University, Malatya, Turkey
| | - Yusuf Tutar
- Division of Biochemistry, Faculty of Pharmacy, Department of Basic Pharmaceutical Sciences, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
35
|
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
|
36
|
Ulbrich F, Kaufmann KB, Meske A, Lagrèze WA, Augustynik M, Buerkle H, Ramao CC, Biermann J, Goebel U. The CORM ALF-186 Mediates Anti-Apoptotic Signaling via an Activation of the p38 MAPK after Ischemia and Reperfusion Injury in Retinal Ganglion Cells. PLoS One 2016; 11:e0165182. [PMID: 27764224 PMCID: PMC5072679 DOI: 10.1371/journal.pone.0165182] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/08/2016] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Ischemia and reperfusion injury may induce apoptosis and lead to sustained tissue damage and loss of function, especially in neuronal organs. While carbon monoxide is known to exert protective effects after various harmful events, the mechanism of carbon monoxide releasing molecules in neuronal tissue has not been investigated yet. We hypothesize that the carbon monoxide releasing molecule (CORM) ALF-186, administered after neuronal ischemia-reperfusion injury (IRI), counteracts retinal apoptosis and its involved signaling pathways and consecutively reduces neuronal tissue damage. METHODS IRI was performed in rat´s retinae for 1 hour. The water-soluble CORM ALF-186 (10 mg/kg) was administered intravenously via a tail vein after reperfusion. After 24 and 48 hours, retinal tissue was harvested to analyze mRNA and protein expression of Bcl-2, Bax, Caspase-3, ERK1/2, p38 and JNK. Densities of fluorogold pre-labeled retinal ganglion cells (RGC) were analyzed 7 days after IRI. Immunohistochemistry was performed on retinal cross sections. RESULTS ALF-186 significantly reduced IRI mediated loss of RGC. ALF-186 treatment differentially affected mitogen-activated protein kinases (MAPK) phosphorylation: ALF-186 activated p38 and suppressed ERK1/2 phosphorylation, while JNK remained unchanged. Furthermore, ALF-186 treatment affected mitochondrial apoptosis, decreasing pro-apoptotic Bax and Caspase-3-cleavage, but increasing anti-apoptotic Bcl-2. Inhibition of p38-MAPK using SB203580 reduced ALF-186 mediated anti-apoptotic effects. CONCLUSION In this study, ALF-186 mediated substantial neuroprotection, affecting intracellular apoptotic signaling, mainly via MAPK p38. CORMs may thus represent a promising therapeutic alternative treating neuronal IRI.
Collapse
Affiliation(s)
- Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kai B. Kaufmann
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Meske
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolf A. Lagrèze
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Augustynik
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carlos C. Ramao
- Instituto de Tecnologia Química e Biológica-António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- Alfama Ltd., Instituto de Biologia Experimental e Tecnológica, IBET, Oeiras, Portugal
| | - Julia Biermann
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
37
|
Ali SI, Alhusseini NF, Atteia HH, Idris RAES, Hasan RA. Renoprotective effect of a combination of garlic and telmisartan against ischemia/reperfusion-induced kidney injury in obese rats. Free Radic Res 2016; 50:966-86. [PMID: 27405440 DOI: 10.1080/10715762.2016.1211644] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity enhances the frequency and severity of acute kidney injury (AKI). Telmisartan pre-treatment was used experimentally in the amelioration of ischemia/reperfusion (IR)-induced AKI. However, there is a lack of evidence regarding its beneficial effects on AKI in obese animals. The present study, therefore, aimed to explore the protective effects of garlic and/or telmisartan against renal damage induced by unilateral IR in obese rats. Meloxicam was used as a standard anti-inflammatory agent. Prophylactic oral administration of meloxicam (3 mg kg(-1)), garlic (500 mg kg(-1)) and/or telmisartan (5 and 10 mg kg(-1)) for 4 wk protected against renal function deterioration induced by IR in obese rats. Both doses of telmisartan significantly reduced serum total cholesterol and triacyglycerol levels as well as peri-renal adipocytes size and renal fibrosis. Renal nuclear factor-kappa B immunoreactivity, tumor necrosis factor-alpha content as well as interleukin-10, adiponectin receptor 1 and macrophages (M1, M2) polarization markers (CD11c, CD206) mRNA expressions were down-regulated in ischemic kidney tissues and white adipose tissues around them by all treatments. Moreover, garlic, telmisartan and their combinations significantly suppressed oxidative stress in renal ischemic tissues. Histological picture was also improved by these treatments. Interestingly, the combinations provided a greater protection than their monotherapy in a dose-dependent manner. We suppose that this combination may be a promising prophylactic regimen for managing AKI in case of obesity. Thus, future experimental and clinical large-scale studies are necessary.
Collapse
Affiliation(s)
- Sousou Ibrahim Ali
- a Department of Biochemistry, Faculty of Pharmacy , Zagazig University , Zagazig , Sharkia Governorate , Egypt
| | | | - Hebatallah Husseini Atteia
- a Department of Biochemistry, Faculty of Pharmacy , Zagazig University , Zagazig , Sharkia Governorate , Egypt
| | - Reham Abd El-Satar Idris
- a Department of Biochemistry, Faculty of Pharmacy , Zagazig University , Zagazig , Sharkia Governorate , Egypt
| | - Rehab Abdallah Hasan
- c Department of Histology, Faculty of Medicine for Girls , Al-Azhar University , Cairo , Egypt
| |
Collapse
|
38
|
Recipient Hyperbilirubinemia May Reduce Ischemia-Reperfusion Injury but Fails to Improve Outcome in Clinical Liver Transplantation. Gastroenterol Res Pract 2016; 2016:6964856. [PMID: 27313607 PMCID: PMC4893452 DOI: 10.1155/2016/6964856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/24/2016] [Indexed: 02/07/2023] Open
Abstract
Background. Exogenous bilirubin may reduce experimental ischemia-reperfusion injury (IRI) due to its antioxidant properties. We studied if early graft exposure to high bilirubin levels in the recipient affects the early IRI and outcomes after liver transplantation (LTx). Methods. In 427 LTx patients, the AUROC curve based on bilirubin and AST at day 1 identified a cutoff of 2.04 mg/dL for the recipient pretransplant bilirubin. Recipients were grouped as having low (group L, n = 152) or high (group H, n = 275) bilirubin. Both groups had similar donor-related variables (age, preservation time, donor BMI > 28, and donor risk index (DRI)). Results. Alanine (ALT) and aspartate (AST) aminotransferase levels were higher in group L at day 1; ALT levels remained higher at day 2 in group L. LTx from high risk donors (DRI > 2) revealed a trend towards lower transaminases during the first two days after transplantation in group H. One month and 1-year patient survival were similar in groups L and H. High preoperative bilirubin did not affect the risk for early graft dysfunction (EGD), death, or graft loss during the first year after transplantation nor the incidence of acute rejection. LTx using donors with DRI > 2 resulted in similar rates of EGD in both groups. Conclusion. Increased bilirubin appears to reduce the early IRI after LTx yet this improvement was insufficient to improve the clinical outcome.
Collapse
|
39
|
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
|
40
|
5-Aminolevulinic acid regulates the inflammatory response and alloimmune reaction. Int Immunopharmacol 2015; 37:71-78. [PMID: 26643355 DOI: 10.1016/j.intimp.2015.11.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
Abstract
5-Aminolevulinic acid (5-ALA) is a naturally occurring amino acid and precursor of heme and protoporphyrin IX (PpIX). Exogenously administrated 5-ALA increases the accumulation of PpIX in tumor cells specifically due to the compromised metabolism of 5-ALA to heme in mitochondria. PpIX emits red fluorescence by the irradiation of blue light and the formation of reactive oxygen species and singlet oxygen. Thus, performing a photodynamic diagnosis (PDD) and photodynamic therapy (PDT) using 5-ALA have given rise to a new strategy for tumor diagnosis and therapy. In addition to the field of tumor therapy, 5-ALA has been implicated in the treatment of inflammatory disease, autoimmune disease and transplantation due to the anti-inflammation and immunoregulation properties that are elicited with the expression of heme oxygenase (HO)-1, an inducible enzyme that catalyzes the rate-limiting step in the oxidative degradation of heme to free iron, biliverdin and carbon monoxide (CO), in combination with sodium ferrous citrate (SFC), because an inhibitor of HO-1 abolishes the effects of 5-ALA. Furthermore, NF-E2-related factor 2 (Nrf2), mitogen-activated protein kinase (MAPK), and heme are involved in the HO-1 expression. Biliverdin and CO are also known to have anti-apoptotic, anti-inflammatory and immunoregulatory functions. We herein review the current use of 5-ALA in inflammatory diseases, transplantation medicine, and tumor therapy.
Collapse
|
41
|
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
|
42
|
Fredenburgh LE, Kraft BD, Hess DR, Harris RS, Wolf MA, Suliman HB, Roggli VL, Davies JD, Winkler T, Stenzler A, Baron RM, Thompson BT, Choi AM, Welty-Wolf KE, Piantadosi CA. Effects of inhaled CO administration on acute lung injury in baboons with pneumococcal pneumonia. Am J Physiol Lung Cell Mol Physiol 2015; 309:L834-46. [PMID: 26320156 DOI: 10.1152/ajplung.00240.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/14/2015] [Indexed: 12/29/2022] Open
Abstract
Inhaled carbon monoxide (CO) gas has therapeutic potential for patients with acute respiratory distress syndrome if a safe, evidence-based dosing strategy and a ventilator-compatible CO delivery system can be developed. In this study, we used a clinically relevant baboon model of Streptococcus pneumoniae pneumonia to 1) test a novel, ventilator-compatible CO delivery system; 2) establish a safe and effective CO dosing regimen; and 3) investigate the local and systemic effects of CO therapy on inflammation and acute lung injury (ALI). Animals were inoculated with S. pneumoniae (10(8)-10(9) CFU) (n = 14) or saline vehicle (n = 5); in a subset with pneumonia (n = 5), we administered low-dose, inhaled CO gas (100-300 ppm × 60-90 min) at 0, 6, 24, and/or 48 h postinoculation and serially measured blood carboxyhemoglobin (COHb) levels. We found that CO inhalation at 200 ppm for 60 min is well tolerated and achieves a COHb of 6-8% with ambient CO levels ≤ 1 ppm. The COHb level measured at 20 min predicted the 60-min COHb level by the Coburn-Forster-Kane equation with high accuracy. Animals given inhaled CO + antibiotics displayed significantly less ALI at 8 days postinoculation compared with antibiotics alone. Inhaled CO was associated with activation of mitochondrial biogenesis in the lung and with augmentation of renal antioxidative programs. These data support the feasibility of safely delivering inhaled CO gas during mechanical ventilation and provide preliminary evidence that CO may accelerate the resolution of ALI in a clinically relevant nonhuman primate pneumonia model.
Collapse
Affiliation(s)
- Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts;
| | - Bryan D Kraft
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Dean R Hess
- Department of Respiratory Care, Massachusetts General Hospital, Boston, Massachusetts; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - R Scott Harris
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Monroe A Wolf
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Victor L Roggli
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - John D Davies
- Department of Respiratory Care, Duke University Medical Center, Durham, North Carolina
| | - Tilo Winkler
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Alex Stenzler
- 12th Man Technologies, Garden Grove, California; and
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Augustine M Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Karen E Welty-Wolf
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Claude A Piantadosi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
43
|
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
|
44
|
Dabrowska-Zamojcin E, Dziedziejko V, Safranow K, Kurzawski M, Domanski L, Pawlik A. Association between the CX3CR1 gene V249I polymorphism and delayed kidney allograft function. Transpl Immunol 2015; 32:172-4. [PMID: 25898802 DOI: 10.1016/j.trim.2015.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/28/2015] [Accepted: 04/14/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Fractalkine is a member of the chemokine family that acts as an adhesion molecule and as an extracellular chemoattractant promoting cellular migration. In this study, we analysed the association between the CX3CR1 gene V249I (rs3732379) SNP and renal allograft function. METHODS The study enrolled 270 Caucasian kidney allograft recipients. The following parameters were recorded in each case: the recipient's age and gender, delayed graft function (DGF) defined as the need for dialysis in the first 7 days after transplantation, occurrence and number of episodes of acute rejection (AR), and chronic allograft dysfunction (CAD). RESULTS Delayed graft function was diagnosed in 39.2% of individuals with the CC genotype, 22.7% with CT and 23.5% of those with the TT genotype. The differences were statistically significant (CC vs. TT+CT: OR = 2.17; 95% CI = 1.28-3.70, p = 0.0042). In multivariate analysis the CC genotype was an independent and significant predictor of higher risk of DGF. The distribution of genotypes and alleles of the CX3CR1 gene polymorphism among patients with and without AR as well as CAD did not differ significantly. CONCLUSIONS The results of this study suggest that the CX3CR1 gene V249I (rs3732379) SNP CC genotype is associated with increased risk of DGF.
Collapse
Affiliation(s)
- Ewa Dabrowska-Zamojcin
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | - Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | - Leszek Domanski
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Powstancow Wlkp. 72, 70-111 Szczecin, Poland.
| |
Collapse
|
45
|
Chang PF, Lin YC, Liu K, Yeh SJ, Ni YH. Heme oxygenase-1 gene promoter polymorphism and the risk of pediatric nonalcoholic fatty liver disease. Int J Obes (Lond) 2015; 39:1236-40. [PMID: 25835554 DOI: 10.1038/ijo.2015.46] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 02/16/2015] [Accepted: 03/29/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Oxidative stress and the insulin-resistant state are thought to be key components in the pathogenesis of pediatric nonalcoholic fatty liver disease (NAFLD). Heme oxygenase (HO) is important in the defense against oxidative stress. This study aimed to assess the association of HO-1 gene promoter polymorphism and insulin resistance with NAFLD among obese children. METHODS A total of 101 obese children aged 6-17 years were recruited. Anthropometric, serum biochemical variables and biomarkers for glucose and insulin metabolism were measured. We screened the allelic frequencies of (GT)n repeats in the HO-1 gene promoter among these obese children. NAFLD was determined through liver ultrasonography. Because the distribution of numbers of (GT)n repeats was bimodal, we divided the alleles into two classes: class S included shorter (27) repeats, and class L included longer (⩾27) repeats. We assessed the effects of the length of (GT)n repeats in HO-1 gene promoter on pediatric NAFLD. RESULTS Of the 101 obese subjects, 27 (26.7%) had NAFLD. The alanine aminotransferase level was higher in patients carrying L alleles (L/L and L/S) than patients with S alleles (S/S) (46.2±49.3 IU|(-1) versus 30.2±20.1 IU|(-1); P=0.027). The significant risk factors for pediatric NAFLD were patients carrying L alleles (L/L and L/S) (odds ratio (OR)=18.84; 95% confidence interval (CI): 1.45-245.22; P=0.025), homeostasis model assessment of insulin resistance (OR=1.40; 95% CI: 1.07-1.83; P=0.014) and age (OR=1.24; 95% CI: 1.03-1.50; P=0.025). CONCLUSION In this hospital-based study, the obese children with longer GT repeats in the HO-1 gene promoter and insulin resistance were susceptible to NAFLD.
Collapse
Affiliation(s)
- P-F Chang
- 1] Department of Pediatrics, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan [2] Department of Healthcare Administration, Oriental Institute of Technology, Pan-Chiao, New Taipei, Taiwan
| | - Y-C Lin
- 1] Department of Pediatrics, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan [2] Department of Healthcare Administration, Oriental Institute of Technology, Pan-Chiao, New Taipei, Taiwan
| | - K Liu
- Department of Pediatrics, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan
| | - S-J Yeh
- 1] Department of Pediatrics, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan [2] Department of Healthcare Administration, Oriental Institute of Technology, Pan-Chiao, New Taipei, Taiwan
| | - Y-H Ni
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
46
|
Zaza G, Ferraro PM, Tessari G, Sandrini S, Scolari MP, Capelli I, Minetti E, Gesualdo L, Girolomoni G, Gambaro G, Lupo A, Boschiero L. Predictive model for delayed graft function based on easily available pre-renal transplant variables. Intern Emerg Med 2015; 10:135-41. [PMID: 25164408 DOI: 10.1007/s11739-014-1119-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/12/2014] [Indexed: 02/06/2023]
Abstract
Identification of pre-transplant factors influencing delayed graft function (DGF) could have an important clinical impact. This could allow clinicians to early identify dialyzed chronic kidney disease (CKD) patients eligible for special transplant programs, preventive therapeutic strategies and specific post-transplant immunosuppressive treatments. To achieve these objectives, we retrospectively analyzed main demographic and clinical features, follow-up events and outcomes registered in a large dedicated dataset including 2,755 patients compiled collaboratively by four Italian renal/transplant units. The years of transplant ranged from 1984 to 2012. Statistical analysis clearly demonstrated that some recipients' characteristics at the time of transplantation (age and body weight) and dialysis-related variables (modality and duration) were significantly associated with DGF development (p ≤ 0.001). The area under the receiver-operating characteristic (ROC) curve of the final model based on the four identified variables predicting DGF was 0.63 (95 % CI 0.61, 0.65). Additionally, deciles of the score were significantly associated with the incidence of DGF (p value for trend <0.001). Therefore, in conclusion, in our study we identified a pre-operative predictive model for DGF, based on inexpensive and easily available variables, potentially useful in routine clinical practice in most of the Italian and European dialysis units.
Collapse
Affiliation(s)
- Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Fukuda W, Takagi T, Katada K, Mizushima K, Okayama T, Yoshida N, Kamada K, Uchiyama K, Ishikawa T, Handa O, Konishi H, Yagi N, Ichikawa H, Yoshikawa T, Cepinskas G, Naito Y, Itoh Y. Anti-inflammatory effects of carbon monoxide-releasing molecule on trinitrobenzene sulfonic acid-induced colitis in mice. Dig Dis Sci 2014; 59:1142-1151. [PMID: 24442266 DOI: 10.1007/s10620-013-3014-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/20/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIM Recent findings indicate that carbon monoxide (CO) in non-toxic doses exerts a beneficial anti-inflammatory action in various experimental models. However, the precise anti-inflammatory mechanism of CO in the intestine remains unclear. Here, we assessed the effects of a novel water-soluble CO-releasing molecule, CORM-3, on trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. METHODS To induce colitis, C57BL/6 male mice received an enema of TNBS. CORM-3 or its inactive compound, iCORM-3, were administered intraperitoneally, once immediately before, and twice daily after receiving an enema of TNBS. Three days after TNBS administration, the distal colon was removed, assessed for colonic damage and histological scores, polymorphonuclear leukocyte recruitment (tissue-associated myeloperoxidase, MPO activity), and TNF-α, IFN-γ and IL-17A expression (mRNA and protein levels in the colon mucosa). CD4(+) T cells isolated from murine spleens were stimulated with anti-CD3/CD28, in the presence or absence of CORM-3/iCORM-3. The cell supernatants were assessed for TNF-α and IFN-γ expression, 24 h following stimulation. RESULTS Colonic damage and histological scores were significantly increased in TNBS-induced mice compared to sham-operated mice. Tissue-associated MPO activity and expression of TNF-α, IFN-γ, and IL-17A in the colonic mucosa were higher in TNBS-induced colitis mice. The above changes were attenuated in CORM-3-treated mice. Further, CORM-3 was effective in reducing TNF-α and IFN-γ production in anti-CD3/CD28-stimulated CD4(+) T cells. CONCLUSIONS These findings indicate that CO released from CORM-3 ameliorates inflammatory responses in the colon of TNBS-challenged mice at least in part through a mechanism that involves the suppression of inflammatory cell recruitment/activation.
Collapse
Affiliation(s)
- Wataru Fukuda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nakao A, Yamada T, Kohama K, Yoshie N, Fujisaki N, Kotani J. Application of carbon monoxide for treatment of acute kidney injury. Acute Med Surg 2014; 1:127-134. [PMID: 29930836 DOI: 10.1002/ams2.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/16/2014] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury in critically ill patients is common and associated with a substantial increase in morbidity and mortality. Even with aggressive medical care and renal replacement therapy, acute kidney injury remains a significant health care concern. Recent published reports offer new strategies for the prevention and amelioration of acute kidney injury using carbon monoxide. Although considered a toxic environmental gas, carbon monoxide has recently aroused scientific and clinical interest, as its beneficial effects and mechanisms of action have been substantially defined in various in vitro and in vivo experiments. The exogenous application of carbon monoxide can confer cytoprotection by modulating intracellular signaling pathways through its anti-inflammatory, anti-apoptotic, vasodilative, antithrombotic and antiproliferative properties. Thus, evidence is accumulating to support the notion of carbon monoxide treatment for acute kidney disease. In this review, we focus on the extensively analyzed advantageous value of treatment with inhaled/soluble carbon monoxide in the context of kidney injury. Mechanisms such as signaling pathways, as well as an expanded view regarding toxicity and side-effects, are described broadly. In addition, we discuss the clinical applicability of carbon monoxide as a promising therapeutic strategy for the treatment of patients with acute kidney disease based on translating basic experimental findings into clinical application.
Collapse
Affiliation(s)
- Atsunori Nakao
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Taihei Yamada
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Keisuke Kohama
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Norichika Yoshie
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Noritomo Fujisaki
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Joji Kotani
- Department of Emergency, Disaster, and Critical Care Medicine Hyogo College of Medicine Nishinomiya Japan
| |
Collapse
|
49
|
Deng J, Lei C, Chen Y, Fang Z, Yang Q, Zhang H, Cai M, Shi L, Dong H, Xiong L. Neuroprotective gases – Fantasy or reality for clinical use? Prog Neurobiol 2014; 115:210-45. [DOI: 10.1016/j.pneurobio.2014.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 12/17/2022]
|
50
|
Hosick PA, Ahmed EK, Gousset MU, Granger JP, Stec DE. Inhalation of carbon monoxide is ineffective as a long-term therapy to reduce obesity in mice fed a high fat diet. BMC OBESITY 2014. [PMID: 26217498 PMCID: PMC4511028 DOI: 10.1186/2052-9538-1-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background Previous studies have demonstrated that induction of heme oxygenase-1 results in weight loss in several rodent models of obesity. However, the specific role of the heme oxygenase-1 metabolite, carbon monoxide (CO), in this response has yet to be established. We recently reported that chronic treatment with CO releasing molecules results in prevention of weight gain in mice fed a high fat diet. In the present study, we sought to determine the effect of chronic CO inhalation on the development and reversal of high fat diet induced obesity. Results CO inhalation at both levels initially resulted in a prevention and reversal of body weight and fat mass over the first 10 weeks of treatment, however, this effect was not sustained. CO inhalation in the prevention groups also had an early effect to lower fasting blood glucose but this effect also was not sustained. Conclusions Our results demonstrate that CO inhalation has a transient effect to prevent and reduce body weight which is not sustained chronically in mice fed a high fat diet. These results suggest that chronic CO inhalation therapy is not an effective treatment to induce long term weight loss.
Collapse
Affiliation(s)
- Peter A Hosick
- Department of Physiology & Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39216 USA
| | - Elhaitham K Ahmed
- Department of Physiology & Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39216 USA
| | - Monette U Gousset
- Department of Physiology & Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39216 USA
| | - Joey P Granger
- Department of Physiology & Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39216 USA
| | - David E Stec
- Department of Physiology & Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39216 USA
| |
Collapse
|