1
|
Andrianova NV, Buyan MI, Brezgunova AA, Cherkesova KS, Zorov DB, Plotnikov EY. Hemorrhagic Shock and Mitochondria: Pathophysiology and Therapeutic Approaches. Int J Mol Sci 2025; 26:1843. [PMID: 40076469 PMCID: PMC11898946 DOI: 10.3390/ijms26051843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Severe injuries and some pathologies associated with massive bleeding, such as maternal hemorrhage, gastrointestinal and perioperative bleeding, and rupture of an aneurysm, often lead to major blood loss and the development of hemorrhagic shock. A sharp decrease in circulating blood volume triggers a vicious cycle of vasoconstriction and coagulopathy leading to ischemia of all internal organs and, in severe decompensated states, ischemia of the brain and heart. The basis of tissue damage and dysfunction in hemorrhagic shock is an interruption in the supply of oxygen and substrates for energy production to the cells, making the mitochondria a source and target of oxidative stress and proapoptotic signaling. Based on these mechanisms, different strategies are proposed to treat the multiple organ failure that occurs in shock. The main direction of such treatment is to provide the cells with a sufficient amount of substrates that utilize oxidative phosphorylation at different stages and increase the efficiency of energy production by the mitochondria. These strategies include restoring the efficiency of mitochondrial complexes, for example, by restoring the nicotinamide adenine dinucleotide (NAD) pool. Another direction is approaches to minimize oxidative stress as well as apoptosis, which are primarily dependent on the mitochondria. There are also a number of other methods to reduce mitochondrial dysfunction and improve the quality of the mitochondrial population. In this review, we consider such strategies for the treatment of hemorrhagic shock and show the promise of therapeutic approaches aimed at restoring the bioenergetic functions of the cell and protecting mitochondria.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Marina I. Buyan
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Anna A. Brezgunova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Kseniia S. Cherkesova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| |
Collapse
|
2
|
Feth M, Hezel F, Gröger M, Hogg M, Zink F, Kress S, Hoffmann A, Calzia E, Wachter U, Radermacher P, Merz T. Metabolic Effects of Sodium Thiosulfate During Resuscitation from Trauma and Hemorrhage in Cigarette-Smoke-Exposed Cystathionine-γ-Lyase Knockout Mice. Biomedicines 2024; 12:2581. [PMID: 39595147 PMCID: PMC11591741 DOI: 10.3390/biomedicines12112581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/26/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Acute and chronic pre-traumatic cigarette smoke exposure increases morbidity and mortality after trauma and hemorrhage. In mice with a genetic deletion of the H2S-producing enzyme cystathione-γ-lyase (CSE-/-), providing exogenous H2S using sodium thiosulfate (Na2S2O3) improved organ function after chest trauma and hemorrhagic shock. Therefore, we evaluated the effect of Na2S2O3 during resuscitation from blunt chest trauma and hemorrhagic shock on CSE-/- mice with pre-traumatic cigarette smoke (CS) exposure. Since H2S is well established as being able to modify energy metabolism, a specific focus was placed on whole-body metabolic pathways and mitochondrial respiratory activity. METHODS Following CS exposure, the CSE-/- mice underwent anesthesia, surgical instrumentation, blunt chest trauma, hemorrhagic shock for over 1 h (target mean arterial pressure (MAP) ≈ 35 ± 5 mmHg), and resuscitation for up to 8 h comprising lung-protective mechanical ventilation, the re-transfusion of shed blood, fluid resuscitation, and continuous i.v. noradrenaline (NoA) to maintain an MAP ≥ 55 mmHg. At the start of the resuscitation, the mice randomly received either i.v. Na2S2O3 (0.45 mg/gbodyweight; n = 14) or the vehicle (NaCl 0.9%; n = 11). In addition to the hemodynamics, lung mechanics, gas exchange, acid-base status, and organ function, we quantified the parameters of carbohydrate, lipid, and protein metabolism using a primed continuous infusion of stable, non-radioactive, isotope-labeled substrates (gas chromatography/mass spectrometry) and the post-mortem tissue mitochondrial respiratory activity ("high-resolution respirometry"). RESULTS While the hemodynamics and NoA infusion rates did not differ, Na2S2O3 was associated with a trend towards lower static lung compliance (p = 0.071) and arterial PO2 (p = 0.089) at the end of the experiment. The direct, aerobic glucose oxidation rate was higher (p = 0.041) in the Na2S2O3-treated mice, which resulted in lower glycemia levels (p = 0.050) and a higher whole-body CO2 production rate (p = 0.065). The mitochondrial respiration in the heart, kidney, and liver tissue did not differ. While the kidney function was comparable, the Na2S2O3-treated mice showed a trend towards a shorter survival time (p = 0.068). CONCLUSIONS During resuscitation from blunt chest trauma and hemorrhagic shock in CSE-/- mice with pre-traumatic CS exposure, Na2S2O3 was associated with increased direct, aerobic glucose oxidation, suggesting a switch in energy metabolism towards preferential carbohydrate utilization. Nevertheless, treatment with Na2S2O3 coincided with a trend towards worsened lung mechanics and gas exchange, and, ultimately, shorter survival.
Collapse
Affiliation(s)
- Maximilian Feth
- Department of Anesthesiology, Critical Care, Emergency Medicine and Pain Therapy, German Armed Forces Hospital Ulm, 89081 Ulm, Germany
| | - Felix Hezel
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Michael Gröger
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Melanie Hogg
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Fabian Zink
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Sandra Kress
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Andrea Hoffmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Enrico Calzia
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Ulrich Wachter
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, 89081 Ulm, Germany (U.W.); (P.R.)
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
3
|
Slade L, Deane CS, Szewczyk NJ, Etheridge T, Whiteman M. Hydrogen sulfide supplementation as a potential treatment for primary mitochondrial diseases. Pharmacol Res 2024; 203:107180. [PMID: 38599468 DOI: 10.1016/j.phrs.2024.107180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/06/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Primary mitochondrial diseases (PMD) are amongst the most common inborn errors of metabolism causing fatal outcomes within the first decade of life. With marked heterogeneity in both inheritance patterns and physiological manifestations, these conditions present distinct challenges for targeted drug therapy, where effective therapeutic countermeasures remain elusive within the clinic. Hydrogen sulfide (H2S)-based therapeutics may offer a new option for patient treatment, having been proposed as a conserved mitochondrial substrate and post-translational regulator across species, displaying therapeutic effects in age-related mitochondrial dysfunction and neurodegenerative models of mitochondrial disease. H2S can stimulate mitochondrial respiration at sites downstream of common PMD-defective subunits, augmenting energy production, mitochondrial function and reducing cell death. Here, we highlight the primary signalling mechanisms of H2S in mitochondria relevant for PMD and outline key cytoprotective proteins/pathways amenable to post-translational restoration via H2S-mediated persulfidation. The mechanisms proposed here, combined with the advent of potent mitochondria-targeted sulfide delivery molecules, could provide a framework for H2S as a countermeasure for PMD disease progression.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Colleen S Deane
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Nathaniel J Szewczyk
- Medical Research Council Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom; Ohio Musculoskeletal and Neurologic Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, Greece
| | - Timothy Etheridge
- Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom.
| | - Matthew Whiteman
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK.
| |
Collapse
|
4
|
Yang T, Yang Q, Lai Q, Zhao J, Nie L, Liu S, Yang J, Chu C. AP39 inhibits ferroptosis by inhibiting mitochondrial autophagy through the PINK1/parkin pathway to improve myocardial fibrosis with myocardial infarction. Biomed Pharmacother 2023; 165:115195. [PMID: 37516015 DOI: 10.1016/j.biopha.2023.115195] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Research has revealed the involvement of mitochondrial autophagy and iron death in the pathogenesis of myocardial fibrosis. The objective of this study is to investigate whether the mitochondrial-targeted H2S donor AP39 inhibits mitochondrial autophagy and antagonizes myocardial cell iron death through the PINK1/Parkin pathway, thereby improving myocardial fibrosis in rats with myocardial infarction. EXPERIMENTAL APPROACH A rat model of myocardial infarction was created by intraperitoneal injection of a high dose of isoproterenol, and H9c2 myocardial cells were subjected to hypoxic injury induced by CoCl2. Western blot, RT-PCR, transmission electron microscopy, immunohistochemistry, as well as echocardiography, and studies on isolated hearts were employed. KEY RESULTS In the hearts of rats with myocardial infarction, there was a significant accumulation of interstitial collagen fibers, accompanied by downregulation of CSE protein expression, activation of the PINK1/Parkin signaling pathway, and activation of mitochondrial autophagy. Intervention with AP39 resulted in a significant improvement of the aforementioned changes, which could be reversed by the addition of PAG. Similar results were observed in vitro experiments. Furthermore, the addition of CCCP reversed the antagonistic effect of AP39 on myocardial cell iron death, while the addition of RSL3 reversed the inhibitory effect of AP39 on collagen production in myocardial cells. CONCLUSION AND IMPLICATIONS The mitochondrial-targeted H2S donor AP39 can inhibit mitochondrial autophagy through the PINK1/Parkin pathway, antagonize myocardial cell iron death, and improve myocardial fibrosis in rats with myocardial infarction.
Collapse
Affiliation(s)
- Ting Yang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China; School of Pharmaceutical Science of University of South China, Hengyang 421000, Hunan Province, China
| | - Qi Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Qi Lai
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China; School of Pharmaceutical Science of University of South China, Hengyang 421000, Hunan Province, China
| | - Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Shengquan Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China.
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China.
| |
Collapse
|
5
|
Cottey L, Smith JE, Watts S. Optimisation of mitochondrial function as a novel target for resuscitation in haemorrhagic shock: a systematic review. BMJ Mil Health 2023:e002427. [PMID: 37491136 DOI: 10.1136/military-2023-002427] [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: 04/06/2023] [Accepted: 06/10/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Traumatic injury is one of the leading causes of death worldwide, and despite significant improvements in patient care, survival in the most severely injured patients remains unchanged. There is a crucial need for innovative approaches to improve trauma patient outcomes; this is particularly pertinent in remote or austere environments with prolonged evacuation times to definitive care. Studies suggest that maintenance of cellular homeostasis is a critical component of optimal trauma patient management, and as the cell powerhouse, it is likely that mitochondria play a pivotal role. As a result, therapies that optimise mitochondrial function could be an important future target for the treatment of critically ill trauma patients. METHODS A systematic review of the literature was undertaken in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol to determine the potential role of mitochondria in traumatic injury and haemorrhagic shock (HS) and to identify current evidence for mitochondrial optimisation therapies in trauma. Articles were included if they assessed a mitochondrial targeted therapy in comparison to a control group, used a model of traumatic injury and HS and reported a method to assess mitochondrial function. RESULTS The search returned 918 articles with 37 relevant studies relating to mitochondrial optimisation identified. Included studies exploring a range of therapies with potential utility in traumatic injury and HS. Therapies were categorised into the key mitochondrial pathways impacted following traumatic injury and HS: ATP levels, cell death, oxidative stress and reactive oxygen species. CONCLUSION This systematic review provides an overview of the key cellular functions of the mitochondria following traumatic injury and HS and identifies why mitochondrial optimisation could be a viable and valuable target in optimising outcome in severely injured patients in the future.
Collapse
Affiliation(s)
- Laura Cottey
- Academic Department of Military Emergency Medicine, Royal Centre for Defence Medicine, Birmingham, UK
| | - J E Smith
- Academic Department of Military Emergency Medicine, Royal Centre for Defence Medicine, Birmingham, UK
- Emergency Department, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - S Watts
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, UK
| |
Collapse
|
6
|
Wolfschmitt EM, Hogg M, Vogt JA, Zink F, Wachter U, Hezel F, Zhang X, Hoffmann A, Gröger M, Hartmann C, Gässler H, Datzmann T, Merz T, Hellmann A, Kranz C, Calzia E, Radermacher P, Messerer DAC. The effect of sodium thiosulfate on immune cell metabolism during porcine hemorrhage and resuscitation. Front Immunol 2023; 14:1125594. [PMID: 36911662 PMCID: PMC9996035 DOI: 10.3389/fimmu.2023.1125594] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Sodium thiosulfate (Na2S2O3), an H2S releasing agent, was shown to be organ-protective in experimental hemorrhage. Systemic inflammation activates immune cells, which in turn show cell type-specific metabolic plasticity with modifications of mitochondrial respiratory activity. Since H2S can dose-dependently stimulate or inhibit mitochondrial respiration, we investigated the effect of Na2S2O3 on immune cell metabolism in a blinded, randomized, controlled, long-term, porcine model of hemorrhage and resuscitation. For this purpose, we developed a Bayesian sampling-based model for 13C isotope metabolic flux analysis (MFA) utilizing 1,2-13C2-labeled glucose, 13C6-labeled glucose, and 13C5-labeled glutamine tracers. Methods After 3 h of hemorrhage, anesthetized and surgically instrumented swine underwent resuscitation up to a maximum of 68 h. At 2 h of shock, animals randomly received vehicle or Na2S2O3 (25 mg/kg/h for 2 h, thereafter 100 mg/kg/h until 24 h after shock). At three time points (prior to shock, 24 h post shock and 64 h post shock) peripheral blood mononuclear cells (PBMCs) and granulocytes were isolated from whole blood, and cells were investigated regarding mitochondrial oxygen consumption (high resolution respirometry), reactive oxygen species production (electron spin resonance) and fluxes within the metabolic network (stable isotope-based MFA). Results PBMCs showed significantly higher mitochondrial O2 uptake and lowerO 2 • - production in comparison to granulocytes. We found that in response to Na2S2O3 administration, PBMCs but not granulocytes had an increased mitochondrial oxygen consumption combined with a transient reduction of the citrate synthase flux and an increase of acetyl-CoA channeled into other compartments, e.g., for lipid biogenesis. Conclusion In a porcine model of hemorrhage and resuscitation, Na2S2O3 administration led to increased mitochondrial oxygen consumption combined with stimulation of lipid biogenesis in PBMCs. In contrast, granulocytes remained unaffected. Granulocytes, on the other hand, remained unaffected.O 2 • - concentration in whole blood remained constant during shock and resuscitation, indicating a sufficient anti-oxidative capacity. Overall, our MFA model seems to be is a promising approach for investigating immunometabolism; especially when combined with complementary methods.
Collapse
Affiliation(s)
- Eva-Maria Wolfschmitt
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Melanie Hogg
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Josef Albert Vogt
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Fabian Zink
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Ulrich Wachter
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Felix Hezel
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Xiaomin Zhang
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Andrea Hoffmann
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Michael Gröger
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Clair Hartmann
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
| | - Holger Gässler
- Department of Anaesthesiology, Intensive Care Medicine, Emergency Medicine and Pain Therapy, Federal Armed Forces Hospital Ulm, Ulm, Germany
| | - Thomas Datzmann
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
| | - Tamara Merz
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Andreas Hellmann
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Enrico Calzia
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Ulm, Germany
- Clinic for Anesthesia and Intensive Care, University Hospital Ulm, Ulm, Germany
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
7
|
Lohakul J, Jeayeng S, Chaiprasongsuk A, Torregrossa R, Wood ME, Saelim M, Thangboonjit W, Whiteman M, Panich U. Mitochondria-Targeted Hydrogen Sulfide Delivery Molecules Protect Against UVA-Induced Photoaging in Human Dermal Fibroblasts, and in Mouse Skin In Vivo. Antioxid Redox Signal 2022; 36:1268-1288. [PMID: 34235951 DOI: 10.1089/ars.2020.8255] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aims: Oxidative stress and mitochondrial dysfunction play a role in the process of skin photoaging via activation of matrix metalloproteases (MMPs) and the subsequent degradation of collagen. The activation of nuclear factor E2-related factor 2 (Nrf2), a transcription factor controlling antioxidant and cytoprotective defense systems, might offer a pharmacological approach to prevent skin photoaging. We therefore investigated a pharmacological approach to prevent skin photoaging, and also investigated a protective effect of the novel mitochondria-targeted hydrogen sulfide (H2S) delivery molecules AP39 and AP123, and nontargeted control molecules, on ultraviolet A light (UVA)-induced photoaging in normal human dermal fibroblasts (NHDFs) in vitro and the skin of BALB/c mice in vivo. Results: In NHDFs, AP39 and AP123 (50-200 nM) but not nontargeted controls suppressed UVA (8 J/cm2)-mediated cytotoxicity and induction of MMP-1 activity, preserved cellular bioenergetics, and increased the expression of collagen and nuclear levels of Nrf2. In in vivo experiments, topical application of AP39 or AP123 (0.3-1 μM/cm2; but not nontargeted control molecules) to mouse skin before UVA (60 J/cm2) irradiation prevented skin thickening, MMP induction, collagen loss of oxidative stress markers 8-hydroxy-2'-deoxyguanosine (8-OHdG), increased Nrf2-dependent signaling, as well as increased manganese superoxide dismutase levels and levels of the mitochondrial biogenesis marker peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α). Innovation and Conclusion: Targeting H2S delivery to mitochondria may represent a novel approach for the prevention and treatment of skin photoaging, as well as being useful tools for determining the role of mitochondrial H2S in skin disorders and aging. Antioxid. Redox Signal. 36, 1268-1288.
Collapse
Affiliation(s)
- Jinapath Lohakul
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Saowanee Jeayeng
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anyamanee Chaiprasongsuk
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Mark E Wood
- University of Exeter Medical School, Exeter, United Kingdom
| | - Malinee Saelim
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Weerawon Thangboonjit
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Uraiwan Panich
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Gröger M, Hogg M, Abdelsalam E, Kress S, Hoffmann A, Stahl B, Calzia E, Wachter U, Vogt JA, Wang R, Merz T, Radermacher P, McCook O. Effects of Sodium Thiosulfate During Resuscitation From Trauma-and-Hemorrhage in Cystathionine-γ-Lyase Knockout Mice With Diabetes Type 1. Front Med (Lausanne) 2022; 9:878823. [PMID: 35572988 PMCID: PMC9106371 DOI: 10.3389/fmed.2022.878823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Sodium thiosulfate (STS) is a recognized drug with antioxidant and H2S releasing properties. We recently showed that STS attenuated organ dysfunction and injury during resuscitation from trauma-and-hemorrhage in CSE-ko mice, confirming its previously described organ-protective and anti-inflammatory properties. The role of H2S in diabetes mellitus type 1 (DMT1) is controversial: genetic DMT1 impairs H2S biosynthesis, which has been referred to contribute to endothelial dysfunction and cardiomyopathy. In contrast, development and severity of hyperglycemia in streptozotocin(STZ)-induced DMT1 was attenuated in CSE-ko mice. Therefore, we tested the hypothesis whether STS would also exert organ-protective effects in CSE-ko mice with STZ-induced DMT1, similar to our findings in animals without underlying co-morbidity. Methods Under short-term anesthesia with sevoflurane and analgesia with buprenorphine CSE-ko mice underwent DMT1-induction by single STZ injection (100 μg⋅g-1). Seven days later, animals underwent blast wave-induced blunt chest trauma and surgical instrumentation followed by 1 h of hemorrhagic shock (MAP 35 ± 5 mmHg). Resuscitation comprised re-transfusion of shed blood, lung-protective mechanical ventilation, fluid resuscitation and continuous i.v. norepinephrine together with either i.v. STS (0.45 mg⋅g-1) or vehicle (n = 9 in each group). Lung mechanics, hemodynamics, gas exchange, acid-base status, stable isotope-based metabolism, and visceral organ function were assessed. Blood and organs were collected for analysis of cytokines, chemokines, and immunoblotting. Results Diabetes mellitus type 1 was associated with more severe circulatory shock when compared to our previous study using the same experimental design in CSE-ko mice without co-morbidity. STS did not exert any beneficial therapeutic effect. Most of the parameters measured of the inflammatory response nor the tissue expression of marker proteins of the stress response were affected either. Conclusion In contrast to our previous findings in CSE-ko mice without underlying co-morbidity, STS did not exert any beneficial therapeutic effect in mice with STZ-induced DMT1, possibly due to DMT1-related more severe circulatory shock. This result highlights the translational importance of both integrating standard ICU procedures and investigating underlying co-morbidity in animal models of shock research.
Collapse
Affiliation(s)
- Michael Gröger
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Melanie Hogg
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Essam Abdelsalam
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Sandra Kress
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Andrea Hoffmann
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Bettina Stahl
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Enrico Calzia
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Ulrich Wachter
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Josef A. Vogt
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Rui Wang
- Faculty of Science, York University, Toronto, ON, Canada
| | - Tamara Merz
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Ulm, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| | - Oscar McCook
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Ulm, Germany
| |
Collapse
|
9
|
Merz T, McCook O, Brucker C, Waller C, Calzia E, Radermacher P, Datzmann T. H 2S in Critical Illness-A New Horizon for Sodium Thiosulfate? Biomolecules 2022; 12:543. [PMID: 35454132 PMCID: PMC9029606 DOI: 10.3390/biom12040543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 12/13/2022] Open
Abstract
Ever since the discovery of endogenous H2S and the identification of its cytoprotective properties, efforts have been made to develop strategies to use H2S as a therapeutic agent. The ability of H2S to regulate vascular tone, inflammation, oxidative stress, and apoptosis might be particularly useful in the therapeutic management of critical illness. However, neither the inhalation of gaseous H2S, nor the administration of inorganic H2S-releasing salts or slow-releasing H2S-donors are feasible for clinical use. Na2S2O3 is a clinically approved compound with a good safety profile and is able to release H2S, in particular under hypoxic conditions. Pre-clinical studies show promise for Na2S2O3 in the acute management of critical illness. A current clinical trial is investigating the therapeutic potential for Na2S2O3 in myocardial infarct. Pre-eclampsia and COVID-19 pneumonia might be relevant targets for future clinical trials.
Collapse
Affiliation(s)
- Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (O.M.); (E.C.); (P.R.)
- Clinic for Anesthesiology and Intensive Care, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (O.M.); (E.C.); (P.R.)
| | - Cosima Brucker
- Department of Gynecology and Obstetrics, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany;
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany;
| | - Enrico Calzia
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (O.M.); (E.C.); (P.R.)
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (O.M.); (E.C.); (P.R.)
| | - Thomas Datzmann
- Clinic for Anesthesiology and Intensive Care, Ulm University Medical Center, 89081 Ulm, Germany;
| |
Collapse
|
10
|
Gröger M, Hogg M, Abdelsalam E, Kress S, Hoffmann A, Stahl B, Saub V, Denoix N, McCook O, Calzia E, Wolfschmitt EM, Wachter U, Vogt JA, Wang R, Radermacher P, Merz T, Nussbaum BL. Effects of Sodium Thiosulfate During Resuscitation From Trauma-and-Hemorrhage in Cystathionine Gamma Lyase (CSE) Knockout Mice. Shock 2022; 57:131-139. [PMID: 34172609 DOI: 10.1097/shk.0000000000001828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Sodium thiosulfate (Na2S2O3) is a clinically established drug with antioxidant and sulphide-releasing properties. Na2S2O3 mediated neuro- and cardioprotective effects in ischemia/reperfusion models and anti-inflammatory effects in LPS-induced acute lung injury. Moreover, Na2S2O3 improved lung function during resuscitation from hemorrhagic shock in swine with pre-existing atherosclerosis, characterized by decreased expression of cystathionine γ-lyase (CSE), a major source of hydrogen sulfide (H2S) synthesis in the vasculature. Based on these findings, we investigated the effects of Na2S2O3 administration during resuscitation from trauma-and-hemorrhage in mice under conditions of whole body CSE deficit. METHODS After blast wave-induced blunt chest trauma and surgical instrumentation, CSE knockout (CSE-/-) mice underwent 1 h of hemorrhagic shock (MAP 35 ± 5 mm Hg). At the beginning of resuscitation comprising retransfusion, norepinephrine support and lung-protective mechanical ventilation, animals received either i.v. Na2S2O3 (0.45 mg g-1, n = 12) or vehicle (saline, n = 13). Hemodynamics, acid-base status, metabolism using stable isotopes, and visceral organ function were assessed. Blood and organs were collected for analysis of cytokines, mitochondrial respiratory capacity, and immunoblotting. RESULTS Na2S2O3 treatment improved arterial paO2 (P = 0.03) coinciding with higher lung tissue glucocorticoid receptor expression. Norepinephrine requirements were lower in the Na2S2O3 group (P < 0.05), which was associated with lower endogenous glucose production and higher urine output. Na2S2O3 significantly increased renal tissue IκBα and heme oxygenase-1 expression, whereas it lowered kidney IL-6 and MCP-1 levels. CONCLUSION Na2S2O3 exerted beneficial effects during resuscitation of murine trauma-and-hemorrhage in CSE-/- mice, confirming and extending the previously described organ-protective and anti-inflammatory properties of Na2S2O3. The findings make Na2S2O3 a potentially promising therapeutic option in the context of impaired CSE activity and/or reduced endogenous H2S availability.
Collapse
Affiliation(s)
- Michael Gröger
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Melanie Hogg
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Essam Abdelsalam
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Sandra Kress
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Andrea Hoffmann
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Bettina Stahl
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Veronique Saub
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Nicole Denoix
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
- Psychosomatic Medicine and Psychotherapy Clinic, University Hospital Ulm, Germany
| | - Oscar McCook
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Enrico Calzia
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Eva-Maria Wolfschmitt
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Ulrich Wachter
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Germany
| | - Josef A Vogt
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Rui Wang
- Faculty of Science, York University, Toronto, ON, Canada
| | - Peter Radermacher
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Tamara Merz
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
| | - Benedikt L Nussbaum
- Institute for Anaesthesiologic Pathophysiology and Process Engineering, University Hospital Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Germany
| |
Collapse
|
11
|
Affiliation(s)
- Thomas Datzmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Csaba Szabo
- Chair of Pharmacology, OMI Department, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| |
Collapse
|
12
|
Merz T, McCook O, Denoix N, Radermacher P, Waller C, Kapapa T. Biological Connection of Psychological Stress and Polytrauma under Intensive Care: The Role of Oxytocin and Hydrogen Sulfide. Int J Mol Sci 2021; 22:9192. [PMID: 34502097 PMCID: PMC8430789 DOI: 10.3390/ijms22179192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
This paper explored the potential mediating role of hydrogen sulfide (H2S) and the oxytocin (OT) systems in hemorrhagic shock (HS) and/or traumatic brain injury (TBI). Morbidity and mortality after trauma mainly depend on the presence of HS and/or TBI. Rapid "repayment of the O2 debt" and prevention of brain tissue hypoxia are cornerstones of the management of both HS and TBI. Restoring tissue perfusion, however, generates an ischemia/reperfusion (I/R) injury due to the formation of reactive oxygen (ROS) and nitrogen (RNS) species. Moreover, pre-existing-medical-conditions (PEMC's) can aggravate the occurrence and severity of complications after trauma. In addition to the "classic" chronic diseases (of cardiovascular or metabolic origin), there is growing awareness of psychological PEMC's, e.g., early life stress (ELS) increases the predisposition to develop post-traumatic-stress-disorder (PTSD) and trauma patients with TBI show a significantly higher incidence of PTSD than patients without TBI. In fact, ELS is known to contribute to the developmental origins of cardiovascular disease. The neurotransmitter H2S is not only essential for the neuroendocrine stress response, but is also a promising therapeutic target in the prevention of chronic diseases induced by ELS. The neuroendocrine hormone OT has fundamental importance for brain development and social behavior, and, thus, is implicated in resilience or vulnerability to traumatic events. OT and H2S have been shown to interact in physical and psychological trauma and could, thus, be therapeutic targets to mitigate the acute post-traumatic effects of chronic PEMC's. OT and H2S both share anti-inflammatory, anti-oxidant, and vasoactive properties; through the reperfusion injury salvage kinase (RISK) pathway, where their signaling mechanisms converge, they act via the regulation of nitric oxide (NO).
Collapse
Affiliation(s)
- Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Nicole Denoix
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
- Clinic for Psychosomatic Medicine and Psychotherapy, Medical Center, Ulm University, 89081 Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Medical Center, Ulm University, Helmholtzstraße 8/1, 89081 Ulm, Germany; (T.M.); (N.D.); (P.R.)
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90471 Nuremberg, Germany;
| | - Thomas Kapapa
- Clinic for Neurosurgery, Medical Center, Ulm University, 89081 Ulm, Germany;
| |
Collapse
|
13
|
Hydrogen Sulfide and the Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:99-128. [PMID: 34302690 DOI: 10.1007/978-981-16-0991-6_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) is the "third gasotransmitter" recognized alongside nitric oxide (NO) and carbon monoxide (CO). H2S exhibits an array of biological effects in mammalian cells as revealed by studies showing important roles in the cardiovascular system, in cell signalling processes, post-translational modifications and in the immune system. Regarding the latter, using pharmacological and genetic approaches scientists have shown this molecule to have both pro- and anti-inflammatory effects in mammalian systems. The anti-inflammatory effects of H2S appeared to be due to its inhibitory action on the nuclear factor kappa beta signalling pathway; NF-kB representing a transcription factor involved in the regulation pro-inflammatory mediators like nitric oxide, prostaglandins, and cytokines. In contrast, results from several animal model describe a more complicated picture and report on pro-inflammatory effects linked to exposure to this molecule; linked to dosage used and point of administration of this molecule. Overall, roles for H2S in several inflammatory diseases spanning arthritis, atherosclerosis, sepsis, and asthma have been described by researchers. In light this work fascinating research, this chapter will cover H2S biology and its many roles in the immune system.
Collapse
|
14
|
Yu Y, Ye SM, Liu DY, Yang LQ. AP39 ameliorates high fat diet-induced liver injury in young rats via alleviation of oxidative stress and mitochondrial impairment. Exp Anim 2021; 70:553-562. [PMID: 34305077 PMCID: PMC8614011 DOI: 10.1538/expanim.21-0056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complication of childhood obesity and an oxidative stress-related multisystem disease. A mitochondria-targeting hydrogen sulfide
(H2S) donor AP39 has antioxidant property, while the mechanism underlying the function of AP39 on pediatric NAFLD remains undefined. Here, 3-week-old SD rats were received a
high-fat diet (HFD) feeding and injected with AP39 (0.05 or 0.1 mg/kg/day) via the tail vein for up to 7 weeks. AP39 reduced weight gain of HFD rats and improved HFD-caused liver injury, as
evidenced by reduced liver index, improved liver pathological damage, decreased NAFLD activity score, as well as low alanine transaminase (ALT) and aspartate transaminase (AST) activities.
AP39 also reduced serum total cholesterol (TC), triglyceride (TG), low-density lipoprotein-cholesterol (LDL-C) concentrations but increased high-density lipoprotein-cholesterol (HDL-C).
Moreover, AP39 prevented reactive oxygen species (ROS) generation, reduced MDA content and increased glutathione (GSH) level and superoxide dismutase (SOD) activity. Furthermore, AP39
increased H2S level, protected mitochondrial DNA (mtDNA), reduced mitochondrial swelling, and restored mitochondrial membrane potential (MMP) alteration. Notably, AP39 diminished
HIF-1α mRNA and protein level, possibly indicating the alleviation in mitochondrial damage. In short, AP39 protects against HFD-induced liver injury in young rats probably through
attenuating lipid accumulation, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yue Yu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University
| | - Shu-Ming Ye
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Anhui Medical University
| | - De-Yun Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University
| | - Li-Qi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University
| |
Collapse
|
15
|
Gulati A, Jain D, Agrawal NR, Rahate P, Choudhuri R, Das S, Dhibar DP, Prabhu M, Haveri S, Agarwal R, Lavhale MS. Resuscitative Effect of Centhaquine (Lyfaquin ®) in Hypovolemic Shock Patients: A Randomized, Multicentric, Controlled Trial. Adv Ther 2021; 38:3223-3265. [PMID: 33970455 PMCID: PMC8189997 DOI: 10.1007/s12325-021-01760-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/22/2021] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Centhaquine (Lyfaquin®) showed significant efficacy as a resuscitative agent in animal models of haemorrhagic shock. Its safety and tolerability were confirmed in healthy human volunteers. In this study, our primary objective was to determine the safety, and the secondary objective was to assess the efficacy of centhaquine in patients with hypovolemic shock. METHODS A prospective, multicentre, randomized phase II study was conducted in male and female patients aged 18-70 years with hypovolemic shock having systolic BP ≤ 90 mmHg. Patients were randomized in a 1:1 ratio to either the control or centhaquine group. The control group received 100 ml of normal saline infusion over 1 h, while the centhaquine group received 0.01 mg/kg of centhaquine in 100 ml normal saline infusion over 1 h. Every patient received standard of care (SOC) and was followed for 28 days. RESULTS Fifty patients were included, and 45 completed the trial: 22 in the control group and 23 in the centhaquine group. The demographics of patients in both groups were comparable. No adverse event related to centhaquine was recorded in the 28-day observation period. The baseline, Injury Scoring System score, haemoglobin, and haematocrit were similar in both groups. However, 91% of the patients in the centhaquine group needed major surgery, whereas only 68% in the control group (p = 0.0526). Twenty-eight-day all-cause mortality was 0/23 in the centhaquine group and 2/22 in the control group. The percent time in ICU and ventilator support was less in the centhaquine group than in the control group. The total amount of vasopressors needed in the first 48 h of resuscitation was lower in the centhaquine group than in the control group (3.12 ± 2.18 vs. 9.39 ± 4.28 mg). An increase in systolic and diastolic BP from baseline through 48 h was more marked in the centhaquine group than in the control group. Compared with the control group, blood lactate level was lower by 1.75 ± 1.07 mmol/l in the centhaquine group on day 3 of resuscitation. Improvements in base deficit, multiple organ dysfunction syndrome (MODS) score and adult respiratory distress syndrome (ARDS) were greater in the centhaquine group than in the control group. CONCLUSION When added to SOC, centhaquine is a well-tolerated and effective resuscitative agent. It improves the clinical outcome of patients with hypovolemic shock. TRIAL REGISTRATION ClinicalTrials.gov identifier number: NCT04056065.
Collapse
Affiliation(s)
- Anil Gulati
- Professor Emeritus, Midwestern University, Downers Grove, IL USA
| | - Dinesh Jain
- Department of Medicine, Dayanand Medical College & Hospital, Civil Lines, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Nilesh Radheshyam Agrawal
- Department of Neurology, New Era Hospital, Near Jalaram Mandir, Queta Colony, Telephone Exchange Chowk, Central Avenue Road, Nagpur, Maharashtra 440008 India
| | - Prashant Rahate
- Seven Star Hospital Jagnade Square, KDK College Road, Nagpur, Maharashtra 440009 India
| | - Rajat Choudhuri
- Department of Anaesthesiology, Institute of Post-Graduate Medical Education and Research and SSKM Hospital, 244 A.J.C. Bose Road, Kolkata, West Bengal 700020 India
| | - Soumen Das
- Department of Surgery, Institute of Post-Graduate Medical Education and Research and SSKM Hospital, 244 A.J.C. Bose Road, Kolkata, West Bengal 700020 India
| | - Deba Prasad Dhibar
- Department of Internal Medicine, Nehru Hospital, Post-Graduate Institute of Medical Education and Research (PGIMER), Sector-12, Chandigarh, 160 012 India
| | - Madhav Prabhu
- Department of Medicine, KLE’s Dr. Prabhakar Kore Hospital and Medical Research Centre, Nehru Nagar, Belgaum, Karnataka 590010 India
| | - Sameer Haveri
- Department of Orthopaedics, KLE’s Dr. Prabhakar Kore Hospital and Medical Research Centre, Nehru Nagar, Belgaum, Karnataka 590010 India
| | - Rohit Agarwal
- Department of Anaesthesiology, ORIANA Hospital, Plot No.: 6, 7, 8 Ravindrapuri Bhelpur, Varanasi, Uttar Pradesh 221005 India
| | | |
Collapse
|
16
|
Gulati A, Choudhuri R, Gupta A, Singh S, Ali SKN, Sidhu GK, Haque PD, Rahate P, Bothra AR, Singh GP, Maheshwari S, Jeswani D, Haveri S, Agarwal A, Agrawal NR. A Multicentric, Randomized, Controlled Phase III Study of Centhaquine (Lyfaquin ®) as a Resuscitative Agent in Hypovolemic Shock Patients. Drugs 2021; 81:1079-1100. [PMID: 34061314 PMCID: PMC8167383 DOI: 10.1007/s40265-021-01547-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 11/30/2022]
Abstract
Introduction Centhaquine (Lyfaquin®) showed significant safety and efficacy in preclinical and clinical phase I and II studies. Methods A prospective, multicentric, randomized phase III study was conducted in patients with hypovolemic shock, systolic blood pressure (SBP) ≤ 90 mmHg, and blood lactate levels ≥ 2 mmol/L. Patients were randomized in a 2:1 ratio to the centhaquine group (n = 71) or the control (saline) group (n = 34). Every patient received standard of care (SOC) and was followed for 28 days. The study drug (normal saline or centhaquine 0.01 mg/kg) was administered in 100 mL of normal saline infusion over 1 h. The primary objectives were to determine changes (mean through 48 h) in SBP, diastolic blood pressure (DBP), blood lactate levels, and base deficit. The secondary objectives included the amount of fluids, blood products, and vasopressors administered in the first 48 h, duration of hospital stay, time in intensive care units, time on ventilator support, change in acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), and the proportion of patients with 28-day all-cause mortality. Results The demographics of patients and baseline vitals in both groups were comparable. The cause of hypovolemic shock was trauma in 29.4 and 47.1% of control group and centhaquine group patients, respectively, and gastroenteritis in 44.1 and 29.4%, respectively. Shock index (SI) and quick sequential organ failure assessment at baseline were similar in the two groups. An equal amount of fluids and blood products were administered in both groups during the first 48 h of resuscitation. A lesser amount of vasopressors was needed in the first 48 h of resuscitation in the centhaquine group. An increase in SBP from baseline was consistently higher up to 48 h (12.9% increase in area under the curve from 0 to 48 h [AUC0–48]) in the centhaquine group than in the control group. A significant increase in pulse pressure (48.1% increase in AUC0–48) in the centhaquine group compared with the control group suggests improved stroke volume due to centhaquine. The SI was significantly lower in the centhaquine group from 1 h (p = 0.032) to 4 h (p = 0.049) of resuscitation. Resuscitation with centhaquine resulted in a significantly greater number of patients with improved blood lactate (control 46.9%; centhaquine 69.3%; p = 0.03) and the base deficit (control 43.7%; centhaquine 69.8%; p = 0.01) than in the control group. ARDS and MODS improved with centhaquine, and an 8.8% absolute reduction in 28-day all-cause mortality was observed in the centhaquine group. Conclusion Centhaquine is an efficacious resuscitative agent for treating hypovolemic shock. The efficacy of centhaquine in distributive shock is being explored. Trial Registration Clinical Trials Registry, India; ctri.icmr.org.in, CTRI/2019/01/017196; clinicaltrials.gov, NCT04045327. Supplementary Information The online version contains supplementary material available at 10.1007/s40265-021-01547-5.
Collapse
Affiliation(s)
- Anil Gulati
- Pharmazz, Inc., 50 West 75th Street, Suite 105, Willowbrook, IL, 60527, USA.
| | - Rajat Choudhuri
- Institute of Postgraduate Medical Education and Research/SSKM Hospital, Kolkata, West Bengal, India
| | - Ajay Gupta
- Chiranjeev Medical Centre, Jhansi, Uttar Pradesh, India
| | - Saurabh Singh
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - S K Noushad Ali
- ACSR Government Medical College and Hospital, Nellore, Andhra Pradesh, India
| | | | | | | | - Aditya R Bothra
- Rahate Surgical Hospital and ICU, Nagpur, Maharashtra, India
| | - Gyan P Singh
- King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Sanjiv Maheshwari
- Jawahar Lal Nehru Medical College and Attached Hospital, Ajmer, Rajasthan, India
| | - Deepak Jeswani
- Criticare Hospital and Research Institute, Nagpur, Maharashtra, India
| | - Sameer Haveri
- KLE's Dr. Prabhakar Kore Hospital and Medical Research Centre, Belgaum, Karnataka, India
| | | | | |
Collapse
|
17
|
Gulati A, Choudhuri R, Gupta A, Singh S, Noushad Ali SK, Sidhu GK, Haque PD, Rahate P, Bothra AR, Singh GP, Maheshwari S, Jeswani D, Haveri S, Agarwal A, Agrawal NR. A multicentric, randomized, controlled phase III study of centhaquine (Lyfaquin ® ) as a resuscitative agent in hypovolemic shock patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2020.07.30.20068114. [PMID: 33173916 PMCID: PMC7654912 DOI: 10.1101/2020.07.30.20068114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
INTRODUCTION Centhaquine (Lyfaquin ® ) showed significant safety and efficacy in preclinical and clinical phase I and II studies. METHODS A prospective, multicentric, randomized phase III study was conducted in patients with hypovolemic shock having systolic blood pressure (SBP) of ≤90 mm Hg and blood lactate levels of ≥2 mmol/L. Patients were randomized in a 2:1 ratio, 71 patients to the centhaquine group and 34 patients to the control (saline) group. Every patient received standard of care (SOC) and was followed for 28 days. The study drug (normal saline or centhaquine (0.01 mg/kg)) was administered in 100 mL of normal saline infusion over 1 hour. The primary objectives were to determine changes (mean through 48 hours) in SBP, diastolic blood pressure (DBP), blood lactate levels, and base deficit. The secondary objectives included the amount of fluids, blood products, vasopressors administered in the first 48 hours, duration of hospital stay, time in ICU, time on the ventilator support, change in patient's Acute Respiratory Distress Syndrome (ARDS), Multiple Organ Dysfunction Syndrome (MODS) scores, and the proportion of patients with 28-day all-cause mortality. RESULTS The demographics of patients and baseline vitals in both groups were comparable. Trauma was the cause of hypovolemic shock in 29.41% of control and 47.06% of centhaquine, gastroenteritis in 44.12% of control, and 29.41% of centhaquine patients. An equal amount of fluids and blood products were administered in both groups during the first 48 hours of resuscitation. A lesser amount of vasopressors was needed in the first 48 hours of resuscitation in the centhaquine group. An increase in SBP from the baseline was consistently higher in the centhaquine group than in the control. A significant increase in pulse pressure in the centhaquine group than the control group suggests improved stroke volume due to centhaquine. The shock index was significantly lower in the centhaquine group than control from 1 hour (p=0.0320) till 4 hours (p=0.0494) of resuscitation. Resuscitation with centhaquine had a significantly greater number of patients with improved blood lactate and the base deficit than the control group. ARDS and MODS improved with centhaquine, and an 8.8% absolute reduction in 28-day all-cause mortality was observed in the centhaquine group. CONCLUSION Centhaquine is a highly efficacious resuscitative agent for treating hypovolemic shock. The efficacy of centhaquine in distributive shock due to sepsis and COVID-19 is being explored. TRIAL REGISTRATION Clinical Trials Registry, India; ctri.icmr.org.in, CTRI/2019/01/017196; clinicaltrials.gov, NCT04045327 . KEY SUMMARY POINTS A multicentric, randomized, controlled trial was conducted to evaluate the efficacy of centhaquine in hypovolemic shock patients.One hundred and five patients were randomized 2:1 to receive centhaquine or saline. Centhaquine was administered at a dose of 0.01 mg/kg in 100 mL saline and infused over 1 hour. The control group received 100 mL of saline over a 1-hour infusion.Centhaquine improved blood pressure, shock index, reduced blood lactate levels, and improved base deficit. Acute Respiratory Distress Syndrome (ARDS) and Multiple Organ Dysfunction Syndrome (MODS) score improved with centhaquine.An 8.8% absolute reduction in 28-day all-cause mortality was observed in the centhaquine group. There were no drug-related adverse events in the study.
Collapse
|
18
|
Trautwein B, Merz T, Denoix N, Szabo C, Calzia E, Radermacher P, McCook O. ΔMST and the Regulation of Cardiac CSE and OTR Expression in Trauma and Hemorrhage. Antioxidants (Basel) 2021; 10:233. [PMID: 33546491 PMCID: PMC7913715 DOI: 10.3390/antiox10020233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Genetic deletion of 3-mercaptopyruvate sulfurtransferase (MST) is known to result in hypertension and cardiac hypertrophy in older mice, and is associated with increased anxiety-like behaviors. Endogenous hydrogen sulfide (H2S) produced by MST in the mitochondria is also known to be involved in physiological and cellular bioenergetics, and its dysfunction associated with depressive behavior and increased cardiovascular morbidity. Interestingly, early life stress has been shown to lead to a significant loss of cystathionine-γ-lyase (CSE) and oxytocin receptor (OTR) expression in the heart. Thus, we were interested in testing the hypothesis of whether genetic MST mutation (ΔMST) would affect cardiac CSE and OTR expression and affect the mitochondrial respiration in a clinically relevant, resuscitated, mouse model of trauma and hemorrhagic shock. In ΔMST mice, we found a reduction of CSE and OTR in both the naive as well as injured state, in contrast to the wild type (wt) controls. Interestingly, the ΔMST showed a different complex IV response to injury than the wt controls, although our claims are based on the non-demonstrated assumption that naive wt and naive ΔMST mice have comparable complex IV activity. Finally, hemorrhagic shock led to a reduction of CSE and OTR, confirming previous results in the injured mouse heart. To date, the exact mechanisms of the cardiac interaction between H2S and OT are not clear, but they point the way to potential cardioprotective therapies.
Collapse
Affiliation(s)
- Britta Trautwein
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Um, Germany; (B.T.); (T.M.); (N.D.); (E.C.); (P.R.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Um, Germany; (B.T.); (T.M.); (N.D.); (E.C.); (P.R.)
| | - Nicole Denoix
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Um, Germany; (B.T.); (T.M.); (N.D.); (E.C.); (P.R.)
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89070 Ulm, Germany
| | - Csaba Szabo
- Department of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
| | - Enrico Calzia
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Um, Germany; (B.T.); (T.M.); (N.D.); (E.C.); (P.R.)
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Um, Germany; (B.T.); (T.M.); (N.D.); (E.C.); (P.R.)
| | - Oscar McCook
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89070 Ulm, Germany
| |
Collapse
|
19
|
Abstract
The translation of preclinical results into successful clinical therapies remains a challenge in sepsis research. One reason for this lack of translation might be the discrepancy between preclinical models and the clinical reality: nonresuscitated young healthy rodents in contrast to elderly comorbid patients in an intensive care unit. We introduce the mouse intensive care unit (MICU) as a concept to address the lack of resuscitation in preclinical studies as one of the limiting issues in translational research. The MICU reflects standard procedures of the clinical intensive care unit: fluid resuscitation, lung-protective mechanical ventilation, and hemodynamic monitoring and management, all tailored to organ- and function-specific targets. Thus, the MICU gives an experimental animal the intermediate possibility of recovery and survival due to "patient" management, which is not reflected in less complex experimental scenarios, which either result in acute survival or death.
Collapse
Affiliation(s)
- Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, Ulm, Germany
| | - Sandra Kress
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, Ulm, Germany
| | - Michael Gröger
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, Ulm, Germany
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
20
|
Karaman Y, Kaya-Yasar Y, Bozkurt TE, Sahin-Erdemli I. Hydrogen sulfide donors prevent lipopolysaccharide-induced airway hyperreactivity in an in vitro model of chronic inflammation in mice. Basic Clin Pharmacol Toxicol 2020; 128:652-660. [PMID: 33369105 DOI: 10.1111/bcpt.13551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 11/24/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
We aimed to investigate and compare the effects of rapid (NaHS) and slow (GYY4137 and AP39) hydrogen sulfide (H2 S) releasing donors on LPS-induced tracheal hyperreactivity and pro-inflammatory cytokine levels in lung tissues of mice. Tissues were isolated from male BALB/c mice and incubated with LPS (10 µg/mL) in tissue culture. The subgroups were incubated with NaHS, GYY4137 and mitochondria-targeted donor AP39. LPS incubation did not alter contraction response to carbachol, but enhanced 5-HT and bradykinin-induced contractions in tracheal rings, and elevated IL-1β, IL-6 and TNF-α levels in lung homogenates. NaHS at 300 µmol/L and 1000 µmol/L, GYY4137 at 30 µmol/L and 100 µmol/L, and AP39 at 30 nmol/L concentrations inhibited the tracheal hyperreactivity to 5-HT, whereas none of these donors affected the enhanced contraction to bradykinin. GYY4137 was also effective to inhibit 5-HT hyperreactivity acutely. In lung tissues, NaHS prevented the elevation of IL-1β level at 1000 μmol/L, and IL-6 and TNF-α levels at 100 μmol/L concentrations. Incubation with GYY4137 (100 µmol/L) and AP39 (30 nmol/L and 300 nmol/L) inhibited the increase in IL-6 and TNF-α levels, but not IL-1β at concentrations that they affected tracheal hyperreactivity. These results indicate that H2 S donors can decrease inflammation and prevent airway hyperreactivity.
Collapse
Affiliation(s)
- Yasemin Karaman
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| | - Yesim Kaya-Yasar
- Faculty of Pharmacy, Department of Pharmacology, Karadeniz Technical University, Trabzon, Turkey
| | - T Emrah Bozkurt
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| | - Inci Sahin-Erdemli
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
21
|
Abstract
This review addresses the plausibility of hydrogen sulfide (H2S) therapy for acute lung injury (ALI) and circulatory shock, by contrasting the promising preclinical results to the present clinical reality. The review discusses how the narrow therapeutic window and width, and potentially toxic effects, the route, dosing, and timing of administration all have to be balanced out very carefully. The development of standardized methods to determine in vitro and in vivo H2S concentrations, and the pharmacokinetics and pharmacodynamics of H2S-releasing compounds is a necessity to facilitate the safety of H2S-based therapies. We suggest the potential of exploiting already clinically approved compounds, which are known or unknown H2S donors, as a surrogate strategy.
Collapse
|
22
|
Denoix N, McCook O, Ecker S, Wang R, Waller C, Radermacher P, Merz T. The Interaction of the Endogenous Hydrogen Sulfide and Oxytocin Systems in Fluid Regulation and the Cardiovascular System. Antioxidants (Basel) 2020; 9:E748. [PMID: 32823845 PMCID: PMC7465147 DOI: 10.3390/antiox9080748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this review is to explore the parallel roles and interaction of hydrogen sulfide (H2S) and oxytocin (OT) in cardiovascular regulation and fluid homeostasis. Their interaction has been recently reported to be relevant during physical and psychological trauma. However, literature reports on H2S in physical trauma and OT in psychological trauma are abundant, whereas available information regarding H2S in psychological trauma and OT in physical trauma is much more limited. This review summarizes recent direct and indirect evidence of the interaction of the two systems and their convergence in downstream nitric oxide-dependent signaling pathways during various types of trauma, in an effort to better understand biological correlates of psychosomatic interdependencies.
Collapse
Affiliation(s)
- Nicole Denoix
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Sarah Ecker
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Rui Wang
- Faculty of Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany;
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| |
Collapse
|
23
|
Zhu C, Su Y, Juriasingani S, Zheng H, Veramkovich V, Jiang J, Sener A, Whiteman M, Lacefield J, Nagpal D, Alotaibi F, Liu K, Zheng X. Supplementing preservation solution with mitochondria-targeted H 2 S donor AP39 protects cardiac grafts from prolonged cold ischemia-reperfusion injury in heart transplantation. Am J Transplant 2019; 19:3139-3148. [PMID: 31338943 DOI: 10.1111/ajt.15539] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/01/2019] [Accepted: 07/14/2019] [Indexed: 01/25/2023]
Abstract
Heart transplant has been accepted as the standard treatment for end-stage heart failure. Because of its susceptibility to ischemia-reperfusion injury, the heart can be preserved for only 4 to 6 hours in cold static preservation solutions. Prolonged ischemia time is adversely associated with primary graft function and long-term survival. New strategies to preserve donor hearts are urgently needed. We demonstrate that AP39, a mitochondria-targeting hydrogen sulfide donor, significantly increases cardiomyocyte viability and reduces cell apoptosis/death after cold hypoxia/reoxygenation in vitro. It also decreases gene expression of proinflammatory cytokines and preserves mitochondria function. Using an in vivo murine heart transplant model, we show that preserving donor hearts with AP39-supplemented University of Wisconsin solution (n = 7) significantly protects heart graft function, measured by quantitative ultrasound scan, against prolonged cold ischemia-reperfusion injury (24 hours at 4°C), along with reducing tissue injury and fibrosis. Our study demonstrates that supplementing preservation solution with AP39 protects cardiac grafts from prolonged ischemia, highlighting its therapeutic potential in preventing ischemia-reperfusion injury in heart transplant.
Collapse
Affiliation(s)
- Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China.,Department of Pathology, Western University, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Yale Su
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China.,Department of Pathology, Western University, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Smriti Juriasingani
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Hao Zheng
- Department of Pathology, Western University, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Vitali Veramkovich
- Department of Pathology, Western University, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada
| | - Alp Sener
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada.,Department of Surgery, Western University, Ontario, Canada.,Lawson Health Research Institute, Ontario, Canada.,Department of Oncology, Western University, Ontario, Canada
| | - Matthew Whiteman
- University of Exeter Medical School, St. Luke's Campus, Exeter, UK
| | - James Lacefield
- Department of Medical Biophysics, Western University, Ontario, Canada.,Department of Electrical & Computer Engineering, Western University, Ontario, Canada.,Robarts Research Institute, Western University, Ontario, Canada
| | - Dave Nagpal
- Department of Surgery, Western University, Ontario, Canada
| | - Faizah Alotaibi
- Department of Pathology, Western University, Ontario, Canada
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xiufen Zheng
- Department of Pathology, Western University, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Center, Ontario, Canada.,Department of Surgery, Western University, Ontario, Canada.,Lawson Health Research Institute, Ontario, Canada.,Department of Oncology, Western University, Ontario, Canada
| |
Collapse
|
24
|
What's New in Shock, August 2019? Shock 2019; 52:143-145. [PMID: 31306304 DOI: 10.1097/shk.0000000000001363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|