1
|
He X, Zou C, Zhang L, Wu P, Yao Y, Dong K, Ren Y, Hu WW, Li Y, Luo H, Ying B, Luo F, Sun X. Advances in Electrochemical Nitrite Reduction toward Nitric Oxide Synthesis for Biomedical Applications. Adv Healthc Mater 2025; 14:e2403468. [PMID: 39865954 DOI: 10.1002/adhm.202403468] [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: 09/11/2024] [Revised: 12/03/2024] [Indexed: 01/28/2025]
Abstract
Nitric oxide (NO) is an essential molecule in biomedicine, recognized for its antibacterial properties, neuronal modulation, and use in inhalation therapies. The effectiveness of NO-based treatments relies on precise control of NO concentrations tailored to specific therapeutic needs. Electrochemical generation of NO (E-NOgen) via nitrite (NO2 -) reduction offers a scalable and efficient route for controlled NO production, while also addressing environmental concerns by reducing NO2 - pollution and maintaining nitrogen cycle balance. Recent developments in catalysts and E-NOgen devices have propelled NO2 - conversion, enabling on-demand NO production. This review provides an overview of NO2 - reduction pathways, with a focus on cutting-edge Fe/Cu-based E-NOgen catalysts, and explores the development of E-NOgen devices for biomedical use. Challenges and future directions for advancing E-NOgen technologies are also discussed.
Collapse
Affiliation(s)
- Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
| | - Chang Zou
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Limei Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Peilin Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kai Dong
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Sichuan, 250014, China
| | - Yuchun Ren
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Han Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Sichuan, 250014, China
| |
Collapse
|
2
|
Marchesi E, Melloni E, Casciano F, Pozza E, Argazzi R, De Risi C, Preti L, Perrone D, Navacchia ML. Evaluation of Anticancer Activity of Nucleoside-Nitric Oxide Photo-Donor Hybrids. Molecules 2024; 29:3383. [PMID: 39064961 PMCID: PMC11279448 DOI: 10.3390/molecules29143383] [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: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Herein, we report the synthesis of a new hybrid compound based on a 2'-deoxyuridine nucleoside conjugated with a NO photo-donor moiety (dU-t-NO) via CuAAC click chemistry. Hybrid dU-t-NO, as well as two previously reported 2'-deoxyadenosine based hybrids (dAdo-S-NO and dAdo-t-NO), were evaluated for their cytotoxic and cytostatic activities in selected cancer cell lines. dAdo-S-NO and dAdo-t-NO hybrids displayed higher activity with respect to dU-t-NO. All hybrids showed effective release of NO in the micromolar range. The photochemical behavior of the newly reported hybrid, dU-t-NO, was studied in the RKO colon carcinoma cell line, whereas the dAdo-t-NO hybrid was tested in both colon carcinoma RKO and hepatocarcinoma Hep 3B2.1-7 cell lines to evaluate the potential effect of NO released upon irradiation on cell viability. A customized irradiation apparatus for in vitro experiments was also designed.
Collapse
Affiliation(s)
- Elena Marchesi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
| | - Elisabetta Melloni
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Elena Pozza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
| | - Roberto Argazzi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy
| | - Carmela De Risi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
| | - Lorenzo Preti
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Daniela Perrone
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Maria Luisa Navacchia
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy
| |
Collapse
|
3
|
Oh Y, Park K, Jung S, Choi M, Kim T, Lee Y, Choi JY, Kim YH, Jung SY, Hong J. Inhalable Nitric Oxide Delivery Systems for Pulmonary Arterial Hypertension Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308936. [PMID: 38054614 DOI: 10.1002/smll.202308936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/24/2023] [Indexed: 12/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe medical condition characterized by elevated blood pressure in the pulmonary arteries. Nitric oxide (NO) is a gaseous signaling molecule with potent vasodilator effects; however, inhaled NO is limited in clinical practice because of the need for tracheal intubation and the toxicity of high NO concentrations. In this study, inhalable NO-releasing microspheres (NO inhalers) are fabricated to deliver nanomolar NO through a nebulizer. Two NO inhalers with distinct porous structures are prepared depending on the molecular weights of NO donors. It is confirmed that pore formation can be controlled by regulating the migration of water molecules from the external aqueous phase to the internal aqueous phase. Notably, open porous NO inhalers (OPNIs) can deliver NO deep into the lungs through a nebulizer. Furthermore, OPNIs exhibit vasodilatory and anti-inflammatory effects via sustained NO release. In conclusion, the findings suggest that OPNIs with highly porous structures have the potential to serve as tools for PAH treatment.
Collapse
Affiliation(s)
- Yoogyeong Oh
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyungtae Park
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sungwon Jung
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Moonhyun Choi
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Taihyun Kim
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yoojin Lee
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jae Young Choi
- Division of Pediatric Cardiology, Department of Pediatrics, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK
| | - Se Yong Jung
- Division of Pediatric Cardiology, Department of Pediatrics, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jinkee Hong
- School of Chemical & Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
4
|
Muenster S, Zarragoikoetxea I, Moscatelli A, Balcells J, Gaudard P, Pouard P, Marczin N, Janssens SP. Inhaled NO at a crossroads in cardiac surgery: current need to improve mechanistic understanding, clinical trial design and scientific evidence. Front Cardiovasc Med 2024; 11:1374635. [PMID: 38646153 PMCID: PMC11027901 DOI: 10.3389/fcvm.2024.1374635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/27/2024] [Indexed: 04/23/2024] Open
Abstract
Inhaled nitric oxide (NO) has been used in pediatric and adult perioperative cardiac intensive care for over three decades. NO is a cellular signaling molecule that induces smooth muscle relaxation in the mammalian vasculature. Inhaled NO has the unique ability to exert its vasodilatory effects in the pulmonary vasculature without any hypotensive side-effects in the systemic circulation. In patients undergoing cardiac surgery, NO has been reported in numerous studies to exert beneficial effects on acutely lowering pulmonary artery pressure and reversing right ventricular dysfunction and/or failure. Yet, various investigations failed to demonstrate significant differences in long-term clinical outcomes. The authors, serving as an advisory board of international experts in the field of inhaled NO within pediatric and adult cardiac surgery, will discuss how the existing scientific evidence can be further improved. We will summarize the basic mechanisms underlying the clinical applications of inhaled NO and how this translates into the mandate for inhaled NO in cardiac surgery. We will move on to the popular use of inhaled NO and will talk about the evidence base of the use of this selective pulmonary vasodilator. This review will elucidate what kind of clinical and biological barriers and gaps in knowledge need to be solved and how this has impacted in the development of clinical trials. The authors will elaborate on how the optimization of inhaled NO therapy, the development of biomarkers to identify the target population and the definition of response can improve the design of future large clinical trials. We will explain why it is mandatory to gain an international consensus for the state of the art of NO therapy far beyond this expert advisory board by including the different major players in the field, such as the different medical societies and the pharma industry to improve our understanding of the real-life effects of inhaled NO in large scale observational studies. The design for future innovative randomized controlled trials on inhaled NO therapy in cardiac surgery, adequately powered and based on enhanced biological phenotyping, will be crucial to eventually provide scientific evidence of its clinical efficacy beyond its beneficial hemodynamic properties.
Collapse
Affiliation(s)
- Stefan Muenster
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Iratxe Zarragoikoetxea
- Department of Anesthesiology and Intensive Care Medicine, Hospital Universitari I Politècnic Fe, Valencia, Spain
| | - Andrea Moscatelli
- Neonatal and Pediatric Intensive Care Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Joan Balcells
- Pediatric Intensive Care Unit, Vall d’Hebron Barcelona Campus Hospitalari, Universitari Vall d'Hebron, Barcelona, Spain
| | - Philippe Gaudard
- Department of Anesthesiology and Critical Care Medicine Arnaud de Villeneuve, CHU Montpellier, University of Montpellier, PhyMedExp, INSERM, CNRS, Montpellier, France
| | - Philippe Pouard
- Department of Anesthesiology and Critical Care, Assistance Publique-Hopitaux de Paris, Hopital Necker-Enfants Malades, Paris, France
| | - Nandor Marczin
- Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Stefan P. Janssens
- Cardiac Intensive Care, Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Adamik B, Frostell C, Dragan B, Paslawska U, Zielinski S, Paslawski R, Janiszewski A, Zielinska M, Ryniak S, Albert J, Gozdzik W. Abnormalities of Coagulation and Fibrinolysis Assessed by Thromboelastometry in an Endotoxic Shock Model in Piglets Treated with Nitric Oxide and Hydrocortisone. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0011. [PMID: 38847555 DOI: 10.2478/aite-2024-0011] [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: 09/05/2023] [Accepted: 04/18/2024] [Indexed: 06/24/2024]
Abstract
This is an animal model study to investigate changes in hemostasis during endotoxemic shock and to determine whether the combination of inhaled nitric oxide (iNO) + intravenous hydrocortisone had an effect on clot formation and fibrinolysis. iNO selectively decreases pulmonary artery pressure, without affecting cardiac index or systemic vascular resistance; however, the results of studies on the possible consequences of iNO administration on coagulation are inconsistent and require further research. Thirty-four piglets were included. Administering endotoxin caused severe hypodynamic shock. Half of the animals received iNO (30 ppm) + hydrocortisone, starting 3 h after endotoxin infusion and continuing to the end of the study. All animals developed coagulation disorders, manifested by a tendency to hypocoagulation; at the same time, fibrinolysis was impaired. Coagulation and fibrinolysis disorders persisted after endotoxin infusion was discontinued, with worse severity in the animals that died before the study was terminated. Administering iNO + hydrocortisone did not cause further changes in coagulation and fibrinolysis parameters, either during or after the endotoxin challenge, suggesting that potential therapeutic interventions with iNO to lower pulmonary arterial pressure will not affect hemostasis.
Collapse
Affiliation(s)
- Barbara Adamik
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland
| | - Claes Frostell
- Department of Anesthesia and Intensive Care, Karolinska Institutet at Danderyd Hospital, Stockholm, Sweden
| | - Barbara Dragan
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland
| | - Urszula Paslawska
- Nicolaus Copernicus University, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Torun, Poland
- Department of Internal Medicine and Clinic of Horses, Dogs and Cats, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Stanislaw Zielinski
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland
| | - Robert Paslawski
- Nicolaus Copernicus University, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Torun, Poland
| | - Adrian Janiszewski
- Department of Internal Disease and Diagnostics, Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Sciences, Poznan, Poland
| | - Marzena Zielinska
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland
| | - Stanislaw Ryniak
- Department of Anesthesia and Intensive Care, Karolinska Institutet at Danderyd Hospital, Stockholm, Sweden
| | - Johanna Albert
- Department of Anesthesia and Intensive Care, Karolinska Institutet at Danderyd Hospital, Stockholm, Sweden
| | - Waldemar Gozdzik
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
6
|
Carrola A, Romão CC, Vieira HLA. Carboxyhemoglobin (COHb): Unavoidable Bystander or Protective Player? Antioxidants (Basel) 2023; 12:1198. [PMID: 37371928 DOI: 10.3390/antiox12061198] [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: 05/07/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Carbon monoxide (CO) is a cytoprotective endogenous gas that is ubiquitously produced by the stress response enzyme heme-oxygenase. Being a gas, CO rapidly diffuses through tissues and binds to hemoglobin (Hb) increasing carboxyhemoglobin (COHb) levels. COHb can be formed in erythrocytes or in plasma from cell-free Hb. Herein, it is discussed as to whether endogenous COHb is an innocuous and inevitable metabolic waste product or not, and it is hypothesized that COHb has a biological role. In the present review, literature data are presented to support this hypothesis based on two main premises: (i) there is no direct correlation between COHb levels and CO toxicity, and (ii) COHb seems to have a direct cytoprotective and antioxidant role in erythrocytes and in hemorrhagic models in vivo. Moreover, CO is also an antioxidant by generating COHb, which protects against the pro-oxidant damaging effects of cell-free Hb. Up to now, COHb has been considered as a sink for both exogenous and endogenous CO generated during CO intoxication or heme metabolism, respectively. Hallmarking COHb as an important molecule with a biological (and eventually beneficial) role is a turning point in CO biology research, namely in CO intoxication and CO cytoprotection.
Collapse
Affiliation(s)
- André Carrola
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Carlos C Romão
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Helena L A Vieira
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
7
|
Wang Z, Jin A, Yang Z, Huang W. Advanced Nitric Oxide Generating Nanomedicine for Therapeutic Applications. ACS NANO 2023; 17:8935-8965. [PMID: 37126728 PMCID: PMC10395262 DOI: 10.1021/acsnano.3c02303] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nitric oxide (NO), a gaseous transmitter extensively present in the human body, regulates vascular relaxation, immune response, inflammation, neurotransmission, and other crucial functions. Nitrite donors have been used clinically to treat angina, heart failure, pulmonary hypertension, and erectile dysfunction. Based on NO's vast biological functions, it further can treat tumors, bacteria/biofilms and other infections, wound healing, eye diseases, and osteoporosis. However, delivering NO is challenging due to uncontrolled blood circulation release and a half-life of under five seconds. With advanced biotechnology and the development of nanomedicine, NO donors packaged with multifunctional nanocarriers by physically embedding or chemically conjugating have been reported to show improved therapeutic efficacy and reduced side effects. Herein, we review and discuss recent applications of NO nanomedicines, their therapeutic mechanisms, and the challenges of NO nanomedicines for future scientific studies and clinical applications. As NO enables the inhibition of the replication of DNA and RNA in infectious microbes, including COVID-19 coronaviruses and malaria parasites, we highlight the potential of NO nanomedicines for antipandemic efforts. This review aims to provide deep insights and practical hints into design strategies and applications of NO nanomedicines.
Collapse
Affiliation(s)
- Zhixiong Wang
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| | - Wei Huang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| |
Collapse
|
8
|
Liao KH, Chan TC, Wu CC, Huang WC, Hsu CW, Chuang HC, Wiratama BS, Chiu WT, Lam C. Association between short-term air pollution exposure and traumatic intracranial hemorrhage: pilot evidence from Taiwan. Front Neurol 2023; 14:1087767. [PMID: 37234787 PMCID: PMC10208221 DOI: 10.3389/fneur.2023.1087767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 04/17/2023] [Indexed: 05/28/2023] Open
Abstract
Introduction The detrimental effects of air pollution on the brain are well established. However, few studies have examined the effect of air pollution on traumatic brain injury (TBI). This pilot study evaluated the association between short-term air pollution exposure and traumatic intracranial hemorrhage (TIH). Methods Hospital data of patients with TBI following road traffic accidents were retrospectively collected from the electronic medical records at five trauma centers in Taiwan between 1 January and 31 December 2017. TIH was employed as an outcome measure. All road accident locations were geocoded, and air quality data were collected from the nearest monitoring stations. Air pollutants were entered into five multivariable models. A sensitivity analysis was performed on patients who are vulnerable to suffering TBI after road accidents, including motorcyclists, bicyclists, and pedestrians. Results Among 730 patients with TBI, 327 had TIH. The ages of ≥65 [odds ratio (OR), 3.24; 95% confidence interval (CI), 1.85-5.70], 45-64 (OR, 2.61; 95% CI, 1.64-4.15), and 25-44 (OR, 1.79; 95% CI, 1.13-2.84) years were identified as significant risk factors in the multivariable analysis. In the best-fit multivariable model, exposure to higher concentrations of particulate matter ≤ 2.5 μm in aerodynamic diameter (PM2.5) was associated with an elevated TIH risk (OR, 1.50; 95% CI, 1.17-1.94). The concentration of nitrogen oxides (NOX) did not increase the risk of TIH (OR, 0.45; 95% CI, 0.32-0.61). After categorizing the air pollution concentration according to quartile, the trend tests in the multivariate model showed that the concentrations of PM2.5 and NOX were significant (p = 0.017 and p < 0.001, respectively). There was a negative borderline significant association between temperature and TIH risk (OR, 0.75; 95% CI, 0.56-1.00, p = 0.05). Notably, the single-vehicle crash was a significant risk factor (OR, 2.11; 95% CI, 1.30-3.42) for TIH. Discussion High PM2.5 concentrations and low temperatures are risk factors for TIH in patients with TBI. High NOX concentrations are associated with a lower TIH risk.
Collapse
Affiliation(s)
- Kuo-Hsing Liao
- Department of Neurosurgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Critical Medicine, Department of Emergency and Critical Care Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurotraumatology and Intensive Care, Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ta-Chien Chan
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Public Health, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Chieh Wu
- Emergency Department, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Cheng Huang
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Emergency Department, Department of Emergency and Critical Care Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Center for Education in Medical Simulation, Taipei Medical University, Taipei, Taiwan
- Department of Education and Humanities in Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Wang Hsu
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Emergency Department, Department of Emergency and Critical Care Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Bayu Satria Wiratama
- Department of Biostatistics, Epidemiology, and Population Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Wen-Ta Chiu
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan
- AHMC Health System, Alhambra, CA, United States
| | - Carlos Lam
- Emergency Department, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
Oza PP, Kashfi K. Utility of NO and H 2S donating platforms in managing COVID-19: Rationale and promise. Nitric Oxide 2022; 128:72-102. [PMID: 36029975 PMCID: PMC9398942 DOI: 10.1016/j.niox.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 01/08/2023]
Abstract
Viral infections are a continuing global burden on the human population, underscored by the ramifications of the COVID-19 pandemic. Current treatment options and supportive therapies for many viral infections are relatively limited, indicating a need for alternative therapeutic approaches. Virus-induced damage occurs through direct infection of host cells and inflammation-related changes. Severe cases of certain viral infections, including COVID-19, can lead to a hyperinflammatory response termed cytokine storm, resulting in extensive endothelial damage, thrombosis, respiratory failure, and death. Therapies targeting these complications are crucial in addition to antiviral therapies. Nitric oxide and hydrogen sulfide are two endogenous gasotransmitters that have emerged as key signaling molecules with a broad range of antiviral actions in addition to having anti-inflammatory properties and protective functions in the vasculature and respiratory system. The enhancement of endogenous nitric oxide and hydrogen sulfide levels thus holds promise for managing both early-stage and later-stage viral infections, including SARS-CoV-2. Using SARS-CoV-2 as a model for similar viral infections, here we explore the current evidence regarding nitric oxide and hydrogen sulfide's use to limit viral infection, resolve inflammation, and reduce vascular and pulmonary damage.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, 10091, USA.
| |
Collapse
|
10
|
Becker-Pelster EM, Hahn MG, Delbeck M, Dietz L, Hüser J, Kopf J, Kraemer T, Marquardt T, Mondritzki T, Nagelschmitz J, Nikkho SM, Pires PV, Tinel H, Weimann G, Wunder F, Sandner P, Schuhmacher J, Stasch JP, Truebel HKF. Inhaled mosliciguat (BAY 1237592): targeting pulmonary vasculature via activating apo-sGC. Respir Res 2022; 23:272. [PMID: 36183104 PMCID: PMC9526466 DOI: 10.1186/s12931-022-02189-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Oxidative stress associated with severe cardiopulmonary diseases leads to impairment in the nitric oxide/soluble guanylate cyclase signaling pathway, shifting native soluble guanylate cyclase toward heme-free apo-soluble guanylate cyclase. Here we describe a new inhaled soluble guanylate cyclase activator to target apo-soluble guanylate cyclase and outline its therapeutic potential. METHODS We aimed to generate a novel soluble guanylate cyclase activator, specifically designed for local inhaled application in the lung. We report the discovery and in vitro and in vivo characterization of the soluble guanylate cyclase activator mosliciguat (BAY 1237592). RESULTS Mosliciguat specifically activates apo-soluble guanylate cyclase leading to improved cardiopulmonary circulation. Lung-selective effects, e.g., reduced pulmonary artery pressure without reduced systemic artery pressure, were seen after inhaled but not after intravenous administration in a thromboxane-induced pulmonary hypertension minipig model. These effects were observed over a broad dose range with a long duration of action and were further enhanced under experimental oxidative stress conditions. In a unilateral broncho-occlusion minipig model, inhaled mosliciguat decreased pulmonary arterial pressure without ventilation/perfusion mismatch. With respect to airway resistance, mosliciguat showed additional beneficial bronchodilatory effects in an acetylcholine-induced rat model. CONCLUSION Inhaled mosliciguat may overcome treatment limitations in patients with pulmonary hypertension by improving pulmonary circulation and airway resistance without systemic exposure or ventilation/perfusion mismatch. Mosliciguat has the potential to become a new therapeutic paradigm, exhibiting a unique mode of action and route of application, and is currently under clinical development in phase Ib for pulmonary hypertension.
Collapse
Affiliation(s)
- Eva M Becker-Pelster
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany.
| | - Michael G Hahn
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Martina Delbeck
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Lisa Dietz
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Jörg Hüser
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Johannes Kopf
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Thomas Kraemer
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Tobias Marquardt
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Thomas Mondritzki
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Fakultät für Gesundheit, University Witten/Herdecke, Witten, Germany
| | - Johannes Nagelschmitz
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Sylvia M Nikkho
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Philippe V Pires
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Allschwil, Switzerland
| | - Hanna Tinel
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Gerrit Weimann
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Frank Wunder
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Peter Sandner
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Department of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Joachim Schuhmacher
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
| | - Johannes-Peter Stasch
- Pharmaceuticals R&D, Pharma Research Center, Bayer AG, Aprather Weg 18A, 42113, Wuppertal, Germany
- Institute of Pharmacy, University Halle-Wittenberg, Halle, Germany
| | | |
Collapse
|
11
|
Nitric Oxide Trickle Drives Heme into Hemoglobin and Muscle Myoglobin. Cells 2022; 11:cells11182838. [PMID: 36139413 PMCID: PMC9496899 DOI: 10.3390/cells11182838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Ever since the days of NO being proclaimed as the “molecule of the year”, the molecular effects of this miracle gas on the globins have remained elusive. While its vasodilatory role in the cardiopulmonary system and the vasculature is well recognized, the molecular underpinnings of the NO–globin axis are incompletely understood. We show, by transwell co-culture of nitric oxide (NO) generating, HEK eNOS/nNOS cells, and K562 erythroid or C2C12 muscle myoblasts, that low doses of NO can effectively insert heme into hemoglobin (Hb) and myoglobin (Mb), making NO not only a vasodilator, but also a globin heme trigger. We found this process to be dependent on the NO flux, occurring at low NO doses and fading at higher doses. This NO-triggered heme insertion occurred into Hb in just 30 min in K562 cells and into muscle Mb in C2C12 myoblasts between 30 min and 1 h, suggesting that the classical effect of NO on upregulation of globin (Hb or Mb) is just not transcriptional, but may involve sufficient translational events where NO can cause heme-downloading into the apo-globins (Hb/Mb). This effect of NO is unexpected and highlights its significance in maintaining globins in its heme-containing holo-form, where such heme insertions might be required in the circulating blood or in the muscle cells to perform spontaneous functions.
Collapse
|
12
|
Xia Z, Zhang C, Guo C, Song B, Hu W, Cui Y, Xue Y, Xia M, Xu D, Zhang S, Fang J. Nanoformulation of a carbon monoxide releasing molecule protects against cyclosporin A-induced nephrotoxicity and renal fibrosis via the suppression of the NLRP3 inflammasome mediated TGF-β/Smad pathway. Acta Biomater 2022; 144:42-53. [PMID: 35304324 DOI: 10.1016/j.actbio.2022.03.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 12/14/2022]
Abstract
Cyclosporin A (CsA) induced nephrotoxicity i.e., renal fibrosis is a critical clinical problem in renal transplant patients, in which chronic inflammatory response is the major cause. Previously, we developed a nano-drug delivery system for carbon monoxide (CO), a multi-functional gaseous molecule with a potent anti-inflammatory effect, i.e., SMA/CORM2, which showed therapeutic potential in several inflammatory disease models. Accordingly, in this study, we explored the potential and usefulness of SMA/CORM2 on CsA induced renal fibrosis. When mice were exposed to CsA for 4 weeks, severe injuries in the kidney as revealed by decreased kidney function and histological examination, and activation of NLRP3 inflammasome, as well as renal fibrosis along with the upregulation of transforming growth factor β (TGFβ)/Smad signaling molecule were observed, whereas SMA/CORM2 (1 mg/kg) treatment remarkably ameliorated the inflammatory injury and fibrosis in the kidney. CO is the major effector molecule of SMA/CORM2 which significantly suppressed the activation of NLRP3 inflammasome, and induced the downregulation of TGFβ/Smad signaling. Inhibition of NLRP3 inflammasome by its inhibitor MCC950 also similarly decreased TGFβ/Smad expression and subsequently improved kidney injury and renal fibrosis, suggesting SMA/CORM2 induced suppression of TGFβ/Smad signaling and renal signaling via an NLRP3 inflammasome-dependent pathway. Compared to native CORM2, SMA/CORM2 exhibited better therapeutic/preventive effects owing to its superior water-solubility and bioavailability. These findings strongly indicated the applicability of SMA/CORM2 as an enhanced permeability and retention (EPR) effect-based nanomedicine for CsA induced renal fibrosis as well as other inflammatory diseases. STATEMENT OF SIGNIFICANCE: Carbon monoxide (CO) is an important gaseous signaling molecule that plays a crucial role in the maintenance of homeostasis. Because of its versatile functions, it exhibits the potential as the target molecule for many diseases, including inflammatory diseases and cancer. The development of stable and disease-targeted delivery systems of CO is thus of interest and importance. Previously we developed a nano micellar CO donor SMA/CORM2 which shows superior bioavailability and therapeutic potential in many inflammatory disease models. We reported here, SMA/CORM2, through controlled release of CO, greatly ameliorated CsA-induced renal fibrosis via suppressing the NLRP3 inflammasome mediated TGF-β/Smad pathway. These findings suggest a new anti-inflammatory mechanism of CO, which also provides a new approach for controlling CsA-induced nephrotoxicity.
Collapse
|
13
|
Simple and sensitive nitric oxide biosensor based on the electrocatalysis of horseradish peroxidase on AuNPs@metal-organic framework composite-modified electrode. Mikrochim Acta 2022; 189:162. [PMID: 35348908 PMCID: PMC8961095 DOI: 10.1007/s00604-022-05268-8] [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: 12/19/2021] [Accepted: 03/08/2022] [Indexed: 11/14/2022]
Abstract
Fe-based metal–organic framework (MIL-101(Fe)) was synthesized through a simple solvothermal synthesis and then used to prepare the AuNPs-decorated MIL-101(Fe) nanocomposite (APPPM(Fe)) by a multi-step layer-by-layer assembly process. Benefited from the porous structure of MIL-101(Fe) and the multilayer assemble process, the loading amount of AuNPs on APPPM(Fe) was enhanced and exhibited a fine biocompatible interface and high conductivity. Through the intense Au–S bond, high loading amount of horseradish peroxidase was immobilized on APPPM(Fe) and the native bioactivity of HRP was kept to realize its direct electrochemistry. From the electrochemical kinetics, the constructed biosensor displayed fast electron transfer and good electrocatalysis activity for the detection of nitric oxide (NO) with wide linear range from 0.033 to 5370 μM and a low detection limit of 0.01 μM (3 σ) as well as fine stability, reproducibility and specificity. According to results of real sample analysis, the proposed electrochemical biosensor offers fast and simple detection of NO in real serum. Therefore, the present strategy definitely provided a potential application prospect in NO clinic detection and disease therapy.
Collapse
|
14
|
Fung C, Z'Graggen WJ, Jakob SM, Gralla J, Haenggi M, Rothen HU, Mordasini P, Lensch M, Söll N, Terpolilli N, Feiler S, Oertel MF, Raabe A, Plesnila N, Takala J, Beck J. Inhaled Nitric Oxide Treatment for Aneurysmal SAH Patients With Delayed Cerebral Ischemia. Front Neurol 2022; 13:817072. [PMID: 35250821 PMCID: PMC8894247 DOI: 10.3389/fneur.2022.817072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
Background We demonstrated experimentally that inhaled nitric oxide (iNO) dilates hypoperfused arterioles, increases tissue perfusion, and improves neurological outcome following subarachnoid hemorrhage (SAH) in mice. We performed a prospective pilot study to evaluate iNO in patients with delayed cerebral ischemia after SAH. Methods SAH patients with delayed cerebral ischemia and hypoperfusion despite conservative treatment were included. iNO was administered at a maximum dose of 40 ppm. The response to iNO was considered positive if: cerebral artery diameter increased by 10% in digital subtraction angiography (DSA), or tissue oxygen partial pressure (PtiO2) increased by > 5 mmHg, or transcranial doppler (TCD) values decreased more than 30 cm/sec, or mean transit time (MTT) decreased below 6.5 secs in CT perfusion (CTP). Patient outcome was assessed at 6 months with the modified Rankin Scale (mRS). Results Seven patients were enrolled between February 2013 and September 2016. Median duration of iNO administration was 23 h. The primary endpoint was reached in all patients (five out of 17 DSA examinations, 19 out of 29 PtiO2 time points, nine out of 26 TCD examinations, three out of five CTP examinations). No adverse events necessitating the cessation of iNO were observed. At 6 months, three patients presented with a mRS score of 0, one patient each with an mRS score of 2 and 3, and two patients had died. Conclusion Administration of iNO in SAH patients is safe. These results call for a larger prospective evaluation.
Collapse
Affiliation(s)
- Christian Fung
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| | - Werner J Z'Graggen
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stephan M Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jan Gralla
- Department of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias Haenggi
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans-Ulrich Rothen
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Pasquale Mordasini
- Department of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michael Lensch
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicole Söll
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicole Terpolilli
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany
- Department of Neurosurgery, Munich University Hospital, Munich, Germany
| | - Sergej Feiler
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus F Oertel
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Andreas Raabe
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany
| | - Jukka Takala
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Qian Y, Kumar R, Chug MK, Massoumi H, Brisbois EJ. Therapeutic Delivery of Nitric Oxide Utilizing Saccharide-Based Materials. ACS APPLIED MATERIALS & INTERFACES 2021; 13:52250-52273. [PMID: 34714640 PMCID: PMC9050970 DOI: 10.1021/acsami.1c10964] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
As a gasotransmitter, nitric oxide (NO) regulates physiological pathways and demonstrates therapeutic effects such as vascular relaxation, anti-inflammation, antiplatelet, antithrombosis, antibacterial, and antiviral properties. However, gaseous NO has high reactivity and a short half-life, so NO delivery and storage are critical questions to be solved. One way is to develop stable NO donors and the other way is to enhance the delivery and storage of NO donors from biomaterials. Most of the researchers studying NO delivery and applications are using synthetic polymeric materials, and they have demonstrated significant therapeutic effects of these NO-releasing polymeric materials on cardiovascular diseases, respiratory disease, bacterial infections, etc. However, some researchers are exploring saccharide-based materials to fulfill the same tasks as their synthetic counterparts while avoiding the concerns of biocompatibility, biodegradability, and sustainability. Saccharide-based materials are abundant in nature and are biocompatible and biodegradable, with wide applications in bioengineering, drug delivery, and therapeutic disease treatments. Saccharide-based materials have been implemented with various NO donors (like S-nitrosothiols and N-diazeniumdiolates) via both chemical and physical methods to deliver NO. These NO-releasing saccharide-based materials have exhibited controlled and sustained NO release and demonstrated biomedical applications in various diseases (respiratory, Crohn's, cardiovascular, etc.), skin or wound applications, antimicrobial treatment, bone regeneration, anticoagulation, as well as agricultural and food packaging. This review aims to highlight the studies in methods and progress in developing saccharide-based NO-releasing materials and investigating their potential applications in biomedical, bioengineering, and disease treatment.
Collapse
Affiliation(s)
- Yun Qian
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Rajnish Kumar
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Manjyot Kaur Chug
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hamed Massoumi
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Elizabeth J Brisbois
- School of Chemical, Materials & Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
16
|
Chou HC, Lo CH, Chang LH, Chiu SJ, Hu TM. Organosilica colloids as nitric oxide carriers: Pharmacokinetics and biocompatibility. Colloids Surf B Biointerfaces 2021; 208:112136. [PMID: 34628305 DOI: 10.1016/j.colsurfb.2021.112136] [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: 06/15/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Nitric oxide (NO) is a potential therapeutic agent for various diseases. However, it is challenging to deliver this unstable, free-radical gaseous molecule in the body. Various nanoparticle-based drug delivery systems have been investigated as promising NO carriers without detailed characterization of their biological fate. The purpose of this study is to investigate the pharmacokinetics and biocompatibility of organosilica-based NO-delivering nanocarriers. Two distinct NO nanoformulations, namely NO-SiNP-1 and NO-SiNP-2, were prepared from a thiol-functionalized organosilane using nanoprecipitation and direct aqueous synthesis, respectively. During the preparation, the thiol group was converted to S-nitrosothiol (SNO) under a nitrosation condition. The final products contain SNO-loaded organosilica particles of similar sizes (~130 nm), but of different morphologies and surface charges (between the two formulations). In the in vitro release kinetics study, NO-SiNP-1 exhibited a much slower NO release rate than NO-SiNP-2 (by 5-fold); therefore, the former is considered as a slow NO releaser, and the latter a fast NO releaser. However, in the rat pharmacokinetic study (IV bolus of 50 μmol/kg), NO-SiNP-1 was rapidly eliminated from the blood (within 20 min); in contrast, NO-SiNP-2 was slowly eliminated with an extended circulation time of 12 h for plasma SNO, along with markedly higher plasma levels of nitrite and nitrate. The two formulations are generally biocompatible. In conclusion, the paper presents contrast biological fates of two organosilica colloidal formulations for nitric oxide delivery.
Collapse
Affiliation(s)
- Hung-Chang Chou
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chih-Hui Lo
- School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Li-Hao Chang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Shih-Jiuan Chiu
- School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.
| | - Teh-Min Hu
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Center for Advanced Pharmaceutics and Drug Delivery Research, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| |
Collapse
|
17
|
Patel JK, Schoenfeld E, Hou W, Singer A, Rakowski E, Ahmad S, Patel R, Parikh PB, Smaldone G. Inhaled nitric oxide in adults with in-hospital cardiac arrest: A feasibility study. Nitric Oxide 2021; 115:30-33. [PMID: 34229057 DOI: 10.1016/j.niox.2021.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/19/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND While inhaled nitric oxide (iNO) has revealed benefit in cardiac arrest in an animal model, no published data has yet demonstrated the impact of iNO in humans with cardiac arrest. METHODS In this pilot study, we administered iNO, along with standard post-resuscitative care, in adults with in-hospital cardiac arrest (IHCA) following achievement of return of spontaneous circulation (ROSC) at an academic tertiary medical center. Patients receiving iNO were compared to age-matched controls with IHCA receiving standard care from an institutional registry. The primary outcome was survival to discharge; secondary outcome was favorable neurologic outcome, defined by a Glasgow Outcome Score of 4 or 5. Propensity-score (PS) matching analysis was performed between patients receiving iNO versus controls. RESULTS Twenty adults with IHCA receiving iNO were compared to 199 controls with IHCA. Similar age, Charlson comorbidity index, and initial rhythm were noted in both groups. Patients receiving iNO had higher rates of survival to discharge compared to controls (35% vs 11%, p < 0.0001) but no difference in favorable neurologic outcome (15% vs 9%, p = 0.39) in the unmatched population. In the PS-matched analysis, patients receiving iNO had higher survival to discharge (35% vs 20%, p = 0.0344) than the control group but no difference in favorable neurologic outcome (15% vs 20%, p = 0.13) were noted between both groups. CONCLUSIONS In this pilot study, iNO was associated with significantly higher rates of survival to discharge but not favorable neurologic outcome among patients with IHCA compared to controls. This benefit was also observed in the PS-matched analysis. A large scale randomized controlled trial comparing standard of care supplemented with iNO to standard of care alone is warranted in patients with cardiac arrest (Funded by Stony Brook University Renaissance School of Medicine, ClinicalTrials.gov number, NCT04134078).
Collapse
Affiliation(s)
- Jignesh K Patel
- Resuscitation Research Group, Division of Pulmonary, Critical Care and Sleep Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA.
| | - Elinor Schoenfeld
- Division of Epidemiology and Biostatistics, Department of Family, Population, and Preventive Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Wei Hou
- Division of Epidemiology and Biostatistics, Department of Family, Population, and Preventive Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Adam Singer
- Department of Emergency Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Ewa Rakowski
- Resuscitation Research Group, Division of Pulmonary, Critical Care and Sleep Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Sahar Ahmad
- Resuscitation Research Group, Division of Pulmonary, Critical Care and Sleep Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Rajeev Patel
- Resuscitation Research Group, Division of Pulmonary, Critical Care and Sleep Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Puja B Parikh
- Division of Cardiology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Gerald Smaldone
- Resuscitation Research Group, Division of Pulmonary, Critical Care and Sleep Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| |
Collapse
|
18
|
Kobayashi J. Nitrite in breast milk: roles in neonatal pathophysiology. Pediatr Res 2021; 90:30-36. [PMID: 33173179 DOI: 10.1038/s41390-020-01247-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 11/09/2022]
Abstract
Dietary nitrate has beneficial effects on health maintenance and prevention of lifestyle-related diseases in adulthood by serving as an alternative source of nitric oxide (NO) through the enterosalivary nitrate-nitrite-NO pathway, particularly when endogenous NO generation is lacking due to vascular endothelial dysfunction. However, this pathway is not developed in the early postnatal period due to a lack of oral commensal nitrate-reducing bacteria and less saliva production than in adults. To compensate for the decrease in nitrite during this period, colostrum contains the highest amount of nitrite compared with transitional, mature, and even artificial milk, suggesting that colostrum plays an important role in tentatively replenishing nitrite, in addition to involving a nutritional aspect, until the enterosalivary nitrate-nitrite-NO pathway is established. Increasing evidence demonstrates that breast milk rich in nitrite can be effective in the prevention of neonatal infections and gastrointestinal diseases such as infantile hypertrophic pyloric stenosis and necrotizing enterocolitis, suggesting that breastfeeding is advantageous for newborns at risk, given the physiological role of nitrite in the early postnatal period. IMPACT: The aim of this review is to discuss the physiological roles of nitrite in breast milk and its implications for neonates. Nitrite in breast milk may compensate for the decrease in nitrite during the early neonatal period until the enterosalivary nitrate-nitrite-nitric oxide pathway is established. Breast milk rich in nitrite may be effective in the prevention of neonatal infections and gastrointestinal diseases by providing nitric oxide bioavailability.
Collapse
Affiliation(s)
- Jun Kobayashi
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Saitama, Japan.
| |
Collapse
|
19
|
Nowaczyk A, Kowalska M, Nowaczyk J, Grześk G. Carbon Monoxide and Nitric Oxide as Examples of the Youngest Class of Transmitters. Int J Mol Sci 2021; 22:ijms22116029. [PMID: 34199647 PMCID: PMC8199767 DOI: 10.3390/ijms22116029] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022] Open
Abstract
The year 2021 is the 100th anniversary of the confirmation of the neurotransmission phenomenon by Otto Loewi. Over the course of the hundred years, about 100 neurotransmitters belonging to many chemical groups have been discovered. In order to celebrate the 100th anniversary of the confirmation of neurotransmitters, we present an overview of the first two endogenous gaseous transmitters i.e., nitric oxide, and carbon monoxide, which are often termed as gasotransmitters.
Collapse
Affiliation(s)
- Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland;
- Correspondence: ; Tel.: +48-52-585-3904
| | - Magdalena Kowalska
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland;
| | - Jacek Nowaczyk
- Department of Physical Chemistry and Physicochemistry of Polymers, Faculty of Chemistry, Nicolaus Copernicus University, 7 Gagarina St., 87-100 Toruń, Poland;
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 75 Ujejskiego St., 85-168 Bydgoszcz, Poland;
| |
Collapse
|
20
|
Jiang S, Dandu C, Geng X. Clinical application of nitric oxide in ischemia and reperfusion injury: A literature review. Brain Circ 2021; 6:248-253. [PMID: 33506147 PMCID: PMC7821808 DOI: 10.4103/bc.bc_69_20] [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: 12/08/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/03/2022] Open
Abstract
Ischemia–reperfusion injury (IRI) is a series of multifactorial cellular events that lead to increased cellular dysfunction after the restoration of oxygen delivery to hypoxic tissue, which can result in acute heart failure and cerebral dysfunction. This injury is severe and would lead to significant morbidity and mortality and poses an important therapeutic challenge for physicians. Nitric oxide (NO) minimizes the deleterious effects of IRI on cells. NO donors, such as organic nitrates and sodium nitroprusside, are used systematically to treat heart failure, angina, and pulmonary hypertension. Inhaled NO gas was approved by the FDA in 1999 to treat hypoxic newborns, and its beneficial ameliorations reach outside the realm of lung disease. This review will summarize the clinical application of NO in IRI.
Collapse
Affiliation(s)
- Shangqian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chaitu Dandu
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
21
|
Abstract
Sudden cardiac arrest is a leading cause of death worldwide. Although the methods of cardiopulmonary resuscitation have been improved, mortality is still unacceptably high, and many survivors suffer from lasting neurological deficits due to the post-cardiac arrest syndrome (PCAS). Pathophysiologically, generalized vascular endothelial dysfunction accompanied by platelet activation and systemic inflammation has been implicated in the pathogenesis of PCAS. Because endothelial-derived nitric oxide (NO) plays a central role in maintaining vascular homeostasis, the role of NO-dependent signaling has been a focus of the intense investigation. Recent preclinical studies showed that therapeutic interventions that increase vascular NO bioavailability may improve outcomes after cardiac arrest complicated with PCAS. In particular, NO inhalation therapy has been shown to improve neurological outcomes and survival in multiple species. Clinical studies examining the safety and efficacy of inhaled NO in patients sustaining PCAS are warranted.
Collapse
|
22
|
Adamik B, Frostell C, Paslawska U, Dragan B, Zielinski S, Paslawski R, Janiszewski A, Zielinska M, Ryniak S, Ledin G, Gozdzik W. Platelet dysfunction in a large-animal model of endotoxic shock; effects of inhaled nitric oxide and low-dose steroid. Nitric Oxide 2021; 108:20-27. [PMID: 33400993 DOI: 10.1016/j.niox.2020.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The role of inhaled nitric oxide in the treatment of shock remains controversial and further translational research is needed. Long-term observation studies using a model of endotoxin-induced shock to assess the effect of inhaled nitric oxide on platelet aggregation have not yet been reported. APPROACH AND RESULTS The tests were carried out in an animal model of shock in two 10-h periods. During the first 10 h, endotoxin was infused and the inhibition of platelet aggregation was evaluated; following the termination of endotoxin infusion, the restoration of platelet aggregation was assessed for 10 h. A total of 30 pigs were used (NO group, N = 14; control, N = 16). In the NO group, nitric oxide inhalation (30 ppm) was started 3 h after endotoxin infusion and continued until the end of the study. Treatment with NO selectively decreased pulmonary artery pressure at 4 (p = 0.002) and 8 h (p = 0.05) of the experiment as compared to the control. Endotoxin significantly reduced platelet aggregation, as indicated by the decreased activity of platelet receptors: ASPI, ADP, collagen, and TRAP during the experiment (p < 0.001). Endotoxin had no significant effect on changes in the response of the receptor after ristocetin stimulation. After stopping endotoxin infusion, a significant restoration of receptor activity was observed for collagen and TRAP, while ASPI and ADP remained partially depressed. Inhaled nitric oxide did not cause additional inhibition of platelet aggregation, either during or after endotoxin challenge. CONCLUSIONS A profound reduction in platelet aggregation was observed during endotoxic shock. After stopping endotoxin infusion a restoration of platelet receptor activity was seen. The inhibition of platelet aggregation induced by endotoxin infusion was not intensified by nitric oxide, indicating there was no harmful effect of inhaled nitric oxide on platelet aggregation.
Collapse
Affiliation(s)
- Barbara Adamik
- Department of the Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland.
| | - Claes Frostell
- Department of Anaesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden.
| | - Urszula Paslawska
- Veterinary Insitute, Nicolaus Copernicus University, Torun, Poland; Department of Internal Medicine and Clinic for Horses, Dogs and Cats, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland.
| | - Barbara Dragan
- Department of the Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland.
| | - Stanislaw Zielinski
- Department of the Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland.
| | - Robert Paslawski
- Veterinary Insitute, Nicolaus Copernicus University, Torun, Poland.
| | - Adrian Janiszewski
- Veterinary Institute, Poznan University of Life Science, Poznan, Poland.
| | - Marzena Zielinska
- Department of the Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland.
| | - Stanislaw Ryniak
- Department of Anaesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden.
| | - Gustaf Ledin
- GHP Stockholm Spine Center AB, Upplands Vasby, Sweden.
| | - Waldemar Gozdzik
- Department of the Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Wroclaw, Poland.
| |
Collapse
|
23
|
Weinstain R, Slanina T, Kand D, Klán P. Visible-to-NIR-Light Activated Release: From Small Molecules to Nanomaterials. Chem Rev 2020; 120:13135-13272. [PMID: 33125209 PMCID: PMC7833475 DOI: 10.1021/acs.chemrev.0c00663] [Citation(s) in RCA: 324] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Photoactivatable (alternatively, photoremovable, photoreleasable, or photocleavable) protecting groups (PPGs), also known as caged or photocaged compounds, are used to enable non-invasive spatiotemporal photochemical control over the release of species of interest. Recent years have seen the development of PPGs activatable by biologically and chemically benign visible and near-infrared (NIR) light. These long-wavelength-absorbing moieties expand the applicability of this powerful method and its accessibility to non-specialist users. This review comprehensively covers organic and transition metal-containing photoactivatable compounds (complexes) that absorb in the visible- and NIR-range to release various leaving groups and gasotransmitters (carbon monoxide, nitric oxide, and hydrogen sulfide). The text also covers visible- and NIR-light-induced photosensitized release using molecular sensitizers, quantum dots, and upconversion and second-harmonic nanoparticles, as well as release via photodynamic (photooxygenation by singlet oxygen) and photothermal effects. Release from photoactivatable polymers, micelles, vesicles, and photoswitches, along with the related emerging field of photopharmacology, is discussed at the end of the review.
Collapse
Affiliation(s)
- Roy Weinstain
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Tomáš Slanina
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czech Republic
| | - Dnyaneshwar Kand
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Petr Klán
- Department
of Chemistry and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
24
|
Pisarenko O, Studneva I. Modulating the Bioactivity of Nitric Oxide as a Therapeutic Strategy in Cardiac Surgery. J Surg Res 2020; 257:178-188. [PMID: 32835951 DOI: 10.1016/j.jss.2020.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/29/2022]
Abstract
Cardiac surgery, including cardioplegic arrest and extracorporeal circulation, causes endothelial dysfunction, which can lead to no-reflow phenomenon and reduction of myocardial pump function. Nitric oxide (NO) deficiency is involved in this pathologic process, thereby providing a fundamental basis for the use of NO replacement therapy. Presently used drugs and additives to cardioplegic and heart preservation solutions are not able to reliably protect endothelial cells and cardiomyocytes from ischemia-reperfusion injury. This review discusses promising NO-releasing compounds of various chemical classes for cardioplegia and reperfusion, which effectively maintain NO homeostasis under experimental conditions, and presents the mechanisms of their action on the cardiovascular system. Incomplete preclinical studies and a lack of toxicity assessment, however, hinder translation of these drug candidates into the clinic. Perspectives for modulation of endothelial function using NO-mediated mechanisms are discussed. They are based on the cardioprotective potential of targeting vascular gap junctions and endothelial ion channels, intracoronary administration of progenitor cells, and endothelial-specific microRNAs. Some of these strategies may provide important therapeutic benefits for human cardiovascular interventions.
Collapse
Affiliation(s)
- Oleg Pisarenko
- National Medical Research Center for Cardiology, Institute of Experimental Cardiology, Moscow, Russian Federation.
| | - Irina Studneva
- National Medical Research Center for Cardiology, Institute of Experimental Cardiology, Moscow, Russian Federation
| |
Collapse
|
25
|
Barnes M, Brisbois EJ. Clinical use of inhaled nitric oxide: Local and systemic applications. Free Radic Biol Med 2020; 152:422-431. [PMID: 31785330 DOI: 10.1016/j.freeradbiomed.2019.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/15/2019] [Accepted: 11/21/2019] [Indexed: 12/26/2022]
Abstract
Upon the FDA approval for inhaled nitric oxide (iNO) in 1999 to treat persistent pulmonary hypertension in neonates, iNO has proven to be a beneficial therapeutic in multiple diseases. We aim to review applications of iNO that have modeled its protective and therapeutic attributes, as well as highlight preliminary studies that could allude to future avenues of use. Numerous publications have reported specific incidences where iNO therapy has proved advantageous, while some applications have potential after further validation. Establishing guidelines on dosing, duration, and defined clinical uses are crucial for the future of iNO. Delivery of iNO has been controlled by a sole distributor, and comes with high cost, and lack of portability. A shift in patents has allowed for new designs for iNO device synthesis, with many new developments of iNO medical devices that will likely change the future of iNO in a medical setting.
Collapse
Affiliation(s)
- Megan Barnes
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Elizabeth J Brisbois
- Department of Materials Science & Engineering, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
26
|
Lafond M, Shekhar H, Panmanee W, Collins SD, Palaniappan A, McDaniel CT, Hassett DJ, Holland CK. Bactericidal Activity of Lipid-Shelled Nitric Oxide-Loaded Microbubbles. Front Pharmacol 2020; 10:1540. [PMID: 32082143 PMCID: PMC7002315 DOI: 10.3389/fphar.2019.01540] [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: 08/29/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
The global pandemic of antibiotic resistance is an ever-burgeoning public health challenge, motivating the development of adjunct bactericidal therapies. Nitric oxide (NO) is a potent bioactive gas that induces a variety of therapeutic effects, including bactericidal and biofilm dispersion properties. The short half-life, high reactivity, and rapid diffusivity of NO make therapeutic delivery challenging. The goal of this work was to characterize NO-loaded microbubbles (MB) stabilized with a lipid shell and to assess the feasibility of antibacterial therapy in vitro. MB were loaded with either NO alone (NO-MB) or with NO and octafluoropropane (NO-OFP-MB) (9:1 v/v and 1:1 v/v). The size distribution and acoustic attenuation coefficient of NO-MB and NO-OFP-MB were measured. Ultrasound-triggered release of the encapsulated gas payload was demonstrated with 3-MHz pulsed Doppler ultrasound. An amperometric microelectrode sensor was used to measure NO concentration released from the MB and compared to an NO-OFP-saturated solution. The effect of NO delivery on the viability of planktonic (free living) Staphylococcus aureus (SA) USA 300, a methicillin-resistant strain, was evaluated in a 96 well-plate format. The co-encapsulation of NO with OFP increased the total volume and attenuation coefficient of MB. The NO-OFP-MB were destroyed with a clinical ultrasound scanner with an output of 2.48 MPa peak negative pressure (in situ MI of 1.34) but maintained their echogenicity when exposed to 0.02 MPa peak negative pressure (in situ MI of 0.01. The NO dose in NO-MB and NO-OFP-MB was more than 2-fold higher than the NO-OFP-saturated solution. Delivery of NO-OFP-MB increased bactericidal efficacy compared to the NO-OFP-saturated solution or air and OFP-loaded MB. These results suggest that encapsulation of NO with OFP in lipid-shelled MB enhances payload delivery. Furthermore, these studies demonstrate the feasibility and limitations of NO-OFP-MB for antibacterial applications.
Collapse
Affiliation(s)
- Maxime Lafond
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
| | - Himanshu Shekhar
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
| | - Warunya Panmanee
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sydney D. Collins
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
| | - Arunkumar Palaniappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
| | - Cameron T. McDaniel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Daniel J. Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christy K. Holland
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
27
|
Spina S, Lei C, Pinciroli R, Berra L. Hemolysis and Kidney Injury in Cardiac Surgery: The Protective Role of Nitric Oxide Therapy. Semin Nephrol 2019; 39:484-495. [DOI: 10.1016/j.semnephrol.2019.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Berrino E, Milazzo L, Micheli L, Vullo D, Angeli A, Bozdag M, Nocentini A, Menicatti M, Bartolucci G, di Cesare Mannelli L, Ghelardini C, Supuran CT, Carta F. Synthesis and Evaluation of Carbonic Anhydrase Inhibitors with Carbon Monoxide Releasing Properties for the Management of Rheumatoid Arthritis. J Med Chem 2019; 62:7233-7249. [PMID: 31287314 DOI: 10.1021/acs.jmedchem.9b00845] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Carbon monoxide (CO) is a gas endogenously produced in humans, reported to exhibit anti-inflammatory and cytoprotective effects at low concentration. In this context, CO releasing molecules (CORMs) are attracting enormous interest. Herein, we report a series of small-molecule hybrids consisting of a carbonic anhydrase (CA; EC 4.2.1.1) inhibitor linked to a CORM tail section (CAI-CORMs). All compounds were screened in vitro for their inhibition activity against the human (h) CA I, II, IV, IX, and XII isoforms. On selected CAI-CORM hybrids, the CO releasing properties were evaluated, along with their pain-relieving effect, in a model of rheumatoid arthritis. One CAI-CORM hybrid (5b) induced a higher pain-relieving effect compared to the one exerted by the single administration of CAI (5a) and CORM (15b) fragments, shedding light on the possibility to enhance the pain relief effect of CA inhibitors inserting a CO releasing moiety on the same molecular scaffold.
Collapse
Affiliation(s)
- Emanuela Berrino
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Lisa Milazzo
- Dipartimento di Chimica "Ugo Schiff" , Università di Firenze , Via della Lastruccia 3-13 , Sesto Fiorentino, 50019 Florence , Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , Viale Gaetano Pieraccini 6 , 50139 Florence , Italy
| | - Daniela Vullo
- Dipartimento di Chimica "Ugo Schiff" , Università di Firenze , Via della Lastruccia 3-13 , Sesto Fiorentino, 50019 Florence , Italy
| | - Andrea Angeli
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Murat Bozdag
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Alessio Nocentini
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Marta Menicatti
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Gianluca Bartolucci
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Lorenzo di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , Viale Gaetano Pieraccini 6 , 50139 Florence , Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , Viale Gaetano Pieraccini 6 , 50139 Florence , Italy
| | - Claudiu T Supuran
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| | - Fabrizio Carta
- NEUROFARBA Deptarment, Sezione di Scienze Farmaceutiche e Nutraceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , Sesto Fiorentino, 50019 Florence , Italy
| |
Collapse
|
29
|
Inhaled nitric oxide prevents systemic and pulmonary vasoconstriction due to hemoglobin-based oxygen carrier infusion: A case report. J Crit Care 2019; 51:213-216. [PMID: 30709560 PMCID: PMC10150649 DOI: 10.1016/j.jcrc.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/02/2018] [Accepted: 04/12/2018] [Indexed: 11/24/2022]
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are used in extreme circumstances to increase hemoglobin concentration and improve oxygen delivery when allogenic red blood cell transfusions are contraindicated or not immediately available. However, HBOC-induced severe pulmonary and systemic vasoconstriction due to peripheral nitric oxide (NO) scavenging has stalled its implementation in clinical practice. We present a case of an 87 year-old patient with acute life-threatening anemia who received HBOC while breathing NO gas. This case shows that inhaled NO allows for the safe use of HBOC infusion by preventing HBOC-induced pulmonary and systemic vasoconstriction.
Collapse
|
30
|
Lautner G, Stringer B, Brisbois EJ, Meyerhoff ME, Schwendeman SP. Controlled light-induced gas phase nitric oxide release from S-nitrosothiol-doped silicone rubber films. Nitric Oxide 2019; 86:31-37. [DOI: 10.1016/j.niox.2019.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/12/2018] [Accepted: 01/31/2019] [Indexed: 11/29/2022]
|
31
|
Inhaled nitric oxide to treat intermediate risk pulmonary embolism: A multicenter randomized controlled trial. Nitric Oxide 2019; 84:60-68. [PMID: 30633959 DOI: 10.1016/j.niox.2019.01.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/21/2018] [Accepted: 01/06/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To test the hypothesis that adjunctive inhaled NO would improve RV function and viability in acute PE. METHODS This was a randomized, placebo-controlled, double blind trial conducted at four academic hospitals. Eligible patients had acute PE without systemic arterial hypotension but had RV dysfunction and a treatment plan of standard anticoagulation. Subjects received either oxygen plus 50 parts per million nitrogen (placebo) or oxygen plus 50 ppm NO for 24 h. The primary composite endpoint required a normal RV on echocardiography and a plasma troponin T concentration <14 pg/mL. The secondary endpoint required a blood brain natriuretic peptide concentration <90 pg/mL and a Borg dyspnea score ≤ 2. The sample size of N = 76 tested if 30% more patients treated with NO would achieve the primary endpoint with 80% power and alpha = 5%. RESULTS We randomized 78 patients and after two withdrawals, 38 were treated per protocol in each group. Patients were well matched for baseline conditions. At 24 h, 5/38 (13%) of patients treated with placebo and 9/38 (24%) of patients treated with NO reached the primary endpoint (P = 0.375). The secondary endpoint was reached in 34% with placebo and 13% of the NO (P = 0.11). In a pre-planned post-hoc analysis, we examined how many patients with RV hypokinesis or dilation at enrollment resolved these abnormalities; 29% more patients treated with NO resolved both abnormalities at 24 h (P = 0.010, Cochrane's Q test). CONCLUSIONS In patients with severe submassive PE, inhaled nitric oxide failed to increase the proportion of patients with a normal troponin and echocardiogram but increased the probability of eliminating RV hypokinesis and dilation on echocardiography. CLINICAL TRIAL REGISTRATION NCT01939301.
Collapse
|
32
|
Nagasaka Y, Fernandez BO, Steinbicker AU, Spagnolli E, Malhotra R, Bloch DB, Bloch KD, Zapol WM, Feelisch M. Pharmacological preconditioning with inhaled nitric oxide (NO): Organ-specific differences in the lifetime of blood and tissue NO metabolites. Nitric Oxide 2018; 80:52-60. [PMID: 30114529 PMCID: PMC6198794 DOI: 10.1016/j.niox.2018.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Endogenous nitric oxide (NO) may contribute to ischemic and anesthetic preconditioning while exogenous NO protects against ischemia-reperfusion (I/R) injury in the heart and other organs. Why those beneficial effects observed in animal models do not always translate into clinical effectiveness remains unclear. To mitigate reperfusion damage a source of NO is required. NO inhalation is known to increase tissue NO metabolites, but little information exists about the lifetime of these species. We therefore sought to investigate the fate of major NO metabolite classes following NO inhalation in mice in vivo. METHODS C57BL/6J mice were exposed to 80 ppm NO for 1 h. NO metabolites were measured in blood (plasma and erythrocytes) and tissues (heart, liver, lung, kidney and brain) immediately after NO exposure and up to 48 h thereafter. Concentrations of S-nitrosothiols, N-nitrosamines and NO-heme products as well as nitrite and nitrate were quantified by gas-phase chemiluminescence and ion chromatography. In separate experiments, mice breathed 80 ppm NO for 1 h prior to cardiac I/R injury (induced by coronary arterial ligation for 1 h, followed by recovery). After sacrifice, the size of the myocardial infarction (MI) and the area at risk (AAR) were measured. RESULTS After NO inhalation, elevated nitroso/nitrosyl levels returned to baseline over the next 24 h, with distinct multi-phasic decay profiles in each compartment. S/N-nitroso compounds and NO-hemoglobin in blood decreased exponentially, but remained above baseline for up to 30min, whereas nitrate was elevated for up to 3hrs after discontinuing NO breathing. Hepatic S/N-nitroso species concentrations remained steady for 30min before dropping exponentially. Nitrate only rose in blood, liver and kidney; nitrite tended to be lower in all organs immediately after NO inhalation but fluctuated considerably in concentration thereafter. NO inhalation before myocardial ischemia decreased the ratio of MI/AAR by 30% vs controls (p = 0.002); only cardiac S-nitrosothiols and NO-hemes were elevated at time of reperfusion onset. CONCLUSIONS Metabolites in blood do not reflect NO metabolite status of any organ. Although NO is rapidly inactivated by hemoglobin-mediated oxidation in the circulation, long-lived tissue metabolites may account for the myocardial preconditioning effects of inhaled NO. NO inhalation may afford similar protection in other organs.
Collapse
Affiliation(s)
- Yasuko Nagasaka
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bernadette O Fernandez
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, UK; Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Andrea U Steinbicker
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, University of Münster, Münster, Germany
| | - Ester Spagnolli
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajeev Malhotra
- Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, UK
| | - Donald B Bloch
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Allergy and Clinical Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kenneth D Bloch
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, UK
| | - Warren M Zapol
- Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Martin Feelisch
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, UK; Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
33
|
Discovery and development of sGC stimulators for the treatment of pulmonary hypertension and rare diseases. Nitric Oxide 2018; 77:88-95. [PMID: 29738821 DOI: 10.1016/j.niox.2018.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/26/2022]
Abstract
The NO/sGC/cGMP signaling cascade plays a pivotal role in regulation of cardiovascular, cardiopulmonary and cardiorenal diseases and impairment of this cascade results in severe pathologies. Therefore, pharmacological interventions, targeting this pathway are promising strategies for treating a variety of diseases. Nitrates, supplementing NO and, PDE5 inhibitors preventing cGMP degradation, are used for angina pectoris treatment and the treatment of pulmonary arterial hypertension (PAH), respectively. More recently, a new class of drugs which directly stimulate the sGC enzyme and trigger NO-independent cGMP production was introduced and termed sGC stimulators. In 2013, the first sGC stimulator, riociguat, was approved for the treatment of PAH and chronic thromboembolic pulmonary hypertension (CTEPH). Since cGMP targets multiple intracellular downstream targets, sGC stimulators have shown - beyond the well characterized vasodilatation - anti-fibrotic, anti-inflammatory and anti-proliferative effects. These additional modes of action might extend the therapeutic potential of this drug class substantially. This review summarizes the NO/sGC/cGMP signaling cascades, the discovery and the mode of action of sGC stimulators. Furthermore, the preclinical evidence and development of riociguat for the treatment of PAH and CTEPH is reviewed. Finally, a summary of the antifibrotic effects of sGC stimulators, especially the most recent finding for skin fibrosis are included which may indicate efficacy in fibrotic diseases like Systemic Sclerosis (SSc).
Collapse
|
34
|
Brücken A, Bleilevens C, Berger P, Nolte K, Gaisa NT, Rossaint R, Marx G, Derwall M, Fries M. Effects of inhaled nitric oxide on outcome after prolonged cardiac arrest in mild therapeutic hypothermia treated rats. Sci Rep 2018; 8:6743. [PMID: 29713000 PMCID: PMC5928159 DOI: 10.1038/s41598-018-25213-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/10/2018] [Indexed: 12/28/2022] Open
Abstract
Guidelines endorse targeted temperature management to reduce neurological sequelae and mortality after cardiac arrest (CA). Additional therapeutic approaches are lacking. Inhaled nitric oxide (iNO) given post systemic ischemia/reperfusion injury improves outcomes. Attenuated inflammation by iNO might be crucial in brain protection. iNO augmented mild therapeutic hypothermia (MTH) may improve outcome after CA exceeding the effect of MTH alone. Following ten minutes of CA and three minutes of cardiopulmonary resuscitation, 20 male Sprague-Dawley rats were randomized to receive MTH at 33 °C for 6hrs or MTH + 20ppm iNO for 5hrs; one group served as normothermic control. During the experiment blood was taken for biochemical evaluation. A neurological deficit score was calculated daily for seven days post CA. On day seven, brains and hearts were harvested for histological evaluation. Treatment groups showed a significant decrease in lactate levels six hours post resuscitation in comparison to controls. TNF-α release was significantly lower in MTH + iNO treated animals only at four hours post ROSC. While only the combination of MTH and iNO improved neurological function in a statistically significant manner in comparison to controls on days 4–7 after CA, there was no significant difference between groups treated with MTH and MTH + iNO.
Collapse
Affiliation(s)
- Anne Brücken
- Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Christian Bleilevens
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Philipp Berger
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Kay Nolte
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Nadine T Gaisa
- Institute of Pathology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Rolf Rossaint
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Matthias Derwall
- Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Michael Fries
- Department of Anaesthesiology, St. Vincenz Hospital Limburg, Auf dem Schafsberg, 65549, Limburg, Germany
| |
Collapse
|
35
|
Ruggiero E, Alonso-de Castro S, Habtemariam A, Salassa L. Upconverting nanoparticles for the near infrared photoactivation of transition metal complexes: new opportunities and challenges in medicinal inorganic photochemistry. Dalton Trans 2018; 45:13012-20. [PMID: 27482656 DOI: 10.1039/c6dt01428c] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The article highlights the emergent use of upconverting nanoparticles as tools for the near infrared photoactivation of transition metal complexes, identifying opportunities and challenges of this approach in the context of medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Emmanuel Ruggiero
- CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, 20009, Spain.
| | | | | | - Luca Salassa
- CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, 20009, Spain. and Kimika Fakultatea, Euskal Herriko Unibertsitatea and Donostia International Physics Center (DIPC), P.K. 1072, Donostia-San Sebastián, 20080, Spain and Ikerbasque, Basque Foundation for Science, Bilbao, 48011, Spain
| |
Collapse
|
36
|
Petit PC, Fine DH, Vásquez GB, Gamero L, Slaughter MS, Dasse KA. The Pathophysiology of Nitrogen Dioxide During Inhaled Nitric Oxide Therapy. ASAIO J 2017; 63:7-13. [PMID: 27556146 DOI: 10.1097/mat.0000000000000425] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Administration of inhaled nitric oxide (NO) with the existing compressed gas delivery systems is associated with unavoidable codelivery of nitrogen dioxide (NO2), an unwanted toxic contaminant that forms when mixed with oxygen. The NO2 is generated when NO is diluted with O2-enriched air before delivery to the patient. When NO2 is inhaled by the patient, it oxidizes protective antioxidants within the epithelial lining fluid (ELF) and triggers extracellular damage in the airways. The reaction of NO2 within the ELF triggers oxidative stress (OS), possibly leading to edema, bronchoconstriction, and a reduced forced expiratory volume in 1 second. Nitrogen dioxide has been shown to have deleterious effects on the airways of high-risk patients including neonates, patients with respiratory and heart failure, and the elderly. Minimizing co-delivery of NO2 for the next generation delivery systems will be a necessity to fully optimize the pulmonary perfusion of NO because of vasodilation, whereas minimizing the negative ventilatory and histopathological effects of NO2 exposure during inhaled NO therapy.
Collapse
Affiliation(s)
- Priscilla C Petit
- From the *Biomedical Research, GeNO LLC, Cocoa, Florida; and †Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky
| | | | | | | | | | | |
Collapse
|
37
|
Muenster S, Lieb WS, Fabry G, Allen KN, Kamat SS, Guy AH, Dordea AC, Teixeira L, Tainsh RE, Yu B, Zhu W, Ashpole NE, Malhotra R, Brouckaert P, Bloch DB, Scherrer-Crosbie M, Stamer WD, Kuehn MH, Pasquale LR, Buys ES. The Ability of Nitric Oxide to Lower Intraocular Pressure Is Dependent on Guanylyl Cyclase. Invest Ophthalmol Vis Sci 2017; 58:4826-4835. [PMID: 28973329 PMCID: PMC5624778 DOI: 10.1167/iovs.17-22168] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose While nitric oxide (NO) donors are emerging as treatments for glaucoma, the mechanism by which NO lowers intraocular pressure (IOP) is unclear. NO activates the enzyme guanylyl cyclase (GC) to produce cyclic guanosine monophosphate. We studied the ocular effects of inhaled and topically applied NO gas in mice and lambs, respectively. Methods IOP and aqueous humor (AqH) outflow were measured in WT and GC-1α subunit null (GC-1−/−) mice. Mice breathed 40 parts per million (ppm) NO in O2 or control gas (N2/O2). We also studied the effect of ocular NO gas exposure (80, 250, 500, and 1000 ppm) on IOP in anesthetized lambs. NO metabolites were measured in AqH and plasma. Results In awake WT mice, breathing NO for 40 minutes lowered IOP from 14.4 ± 1.9 mm Hg to 10.9 ± 1.0 mm Hg (n = 11, P < 0.001). Comparable results were obtained in anesthetized WT mice (n = 10, P < 0.001). In awake or anesthetized GC-1−/− mice, IOP did not change under similar experimental conditions (P ≥ 0.08, n = 20). Breathing NO increased in vivo outflow facility in WT but not GC-1−/− mice (+13.7 ± 14.6% vs. −12.1 ± 9.4%, n = 4 each, P < 0.05). In lambs, ocular exposure to NO lowered IOP in a dose-dependent manner (−0.43 mm Hg/ppm NO; n = 5 with 40 total measurements; P = 0.04) without producing corneal pathology or altering pulmonary and systemic hemodynamics. After ocular NO exposure, NO metabolites were increased in AqH (n = 8, P < 0.001) but not in plasma. Conclusions Breathing NO reduced IOP and increased outflow facility in a GC-dependent manner in mice. Exposure of ovine eyes to NO lowers IOP.
Collapse
Affiliation(s)
- Stefan Muenster
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States.,Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Wolfgang S Lieb
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States.,Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Gregor Fabry
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Kaitlin N Allen
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Shivani S Kamat
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Ann H Guy
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Ana C Dordea
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Leandro Teixeira
- Department of Pathological Science, School of Veterinary Medicine, University of Wisconsin, Wisconsin, United States
| | - Robert E Tainsh
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Binglan Yu
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Wei Zhu
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Nicole E Ashpole
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States.,Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Rajeev Malhotra
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium Department of Molecular Biomedical Research, VIB, Ghent, Belgium
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States.,The Center for Immunology and Inflammatory Diseases and the Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - Marielle Scherrer-Crosbie
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States.,Cardiac Ultrasound Laboratory and Division of Cardiology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States.,Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Louis R Pasquale
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Emmanuel S Buys
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
38
|
Jiang WL, Wei HJ, Guo ZY, Ni YR, Yang HQ, Xie SS. Effects of different-intensity laser acupuncture at two adjacent same-meridian acupoints on nitric oxide and soluble guanylate cyclase releases in human. Microcirculation 2017; 24. [PMID: 28665547 DOI: 10.1111/micc.12390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 06/26/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The aim of this study was to detect the influences of LA at nonacupoint and two adjacent acupoints of pericardium meridian on the releases of NO and sGC in 20 healthy subjects. METHODS Different intensities (12, 24, 48 mW) of infrared laser were used for irradiating Jianshi (PC5), Ximen (PC4) acupoints and nonacupoint for 20, 40 minutes, respectively. Semi-circular tubes were taped to the skin surface and filled with NO-scavenging compound for 20 minutes to capture NO and sGC, which were measured using spectrophotometry in a blinded fashion. RESULTS As the increase in the intensity of LA stimulation, the levels of NO releases over acupoints all were significantly increased, NO releases in nonacupoints following the same treatment only changed slightly, sGC amounts were observably enhanced over acupoints, but did not any change in nonacupoint area. Different intensities of LA treatments can sensitively affect the NO and sGC releases over acupoints. This indicated that LA-induced releases of the NO and sGC were specific to acupoints. CONCLUSIONS This is the first evidence reporting that LA induced significant elevations of NO-sGC releases over acupoints, and the enhanced signal molecules contribute to local circulation, which improves the beneficial effects of the therapy.
Collapse
Affiliation(s)
- Wan-Ling Jiang
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Hua-Jiang Wei
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhou-Yi Guo
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Yi-Rong Ni
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Hong-Qin Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education of China, Fujian Normal University, Fuzhou, Fujian, China
| | - Shu-Sen Xie
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education of China, Fujian Normal University, Fuzhou, Fujian, China
| |
Collapse
|
39
|
Qian Y, Matson JB. Gasotransmitter delivery via self-assembling peptides: Treating diseases with natural signaling gases. Adv Drug Deliv Rev 2017; 110-111:137-156. [PMID: 27374785 DOI: 10.1016/j.addr.2016.06.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 06/16/2016] [Accepted: 06/23/2016] [Indexed: 11/19/2022]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are powerful signaling molecules that play a variety of roles in mammalian biology. Collectively called gasotransmitters, these gases have wide-ranging therapeutic potential, but their clinical use is limited by their gaseous nature, extensive reactivity, short half-life, and systemic toxicity. Strategies for gasotransmitter delivery with control over the duration and location of release are therefore vital for developing effective therapies. An attractive strategy for gasotransmitter delivery is though injectable or implantable gels, which can ideally deliver their payload over a controllable duration and then degrade into benign metabolites. Self-assembling peptide-based gels are well-suited to this purpose due to their tunable mechanical properties, easy chemical modification, and inherent biodegradability. In this review we illustrate the biological roles of NO, CO, and H2S, discuss their therapeutic potential, and highlight recent efforts toward their controlled delivery with a focus on peptide-based delivery systems.
Collapse
Affiliation(s)
- Yun Qian
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - John B Matson
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
40
|
Terpolilli NA, Feiler S, Dienel A, Müller F, Heumos N, Friedrich B, Stover J, Thal S, Schöller K, Plesnila N. Nitric oxide inhalation reduces brain damage, prevents mortality, and improves neurological outcome after subarachnoid hemorrhage by resolving early pial microvasospasms. J Cereb Blood Flow Metab 2016; 36:2096-2107. [PMID: 26661144 PMCID: PMC5363657 DOI: 10.1177/0271678x15605848] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/15/2023]
Abstract
Subarachnoid hemorrhage is a stroke subtype with particularly bad outcome. Recent findings suggest that constrictions of pial arterioles occurring early after hemorrhage may be responsible for cerebral ischemia and - subsequently - unfavorable outcome after subarachnoid hemorrhage. Since we recently hypothesized that the lack of nitric oxide may cause post-hemorrhagic microvasospasms, our aim was to investigate whether inhaled nitric oxide, a treatment paradigm selectively delivering nitric oxide to ischemic microvessels, is able to dilate post-hemorrhagic microvasospasms; thereby improving outcome after experimental subarachnoid hemorrhage. C57BL/6 mice were subjected to experimental SAH. Three hours after subarachnoid hemorrhage pial artery spasms were quantified by intravital microscopy, then mice received inhaled nitric oxide or vehicle. For induction of large artery spasms mice received an intracisternal injection of autologous blood. Inhaled nitric oxide significantly reduced number and severity of subarachnoid hemorrhage-induced post-hemorrhage microvasospasms while only having limited effect on large artery spasms. This resulted in less brain-edema-formation, less hippocampal neuronal loss, lack of mortality, and significantly improved neurological outcome after subarachnoid hemorrhage. This suggests that spasms of pial arterioles play a major role for the outcome after subarachnoid hemorrhage and that lack of nitric oxide is an important mechanism of post-hemorrhagic microvascular dysfunction. Reversing microvascular dysfunction by inhaled nitric oxide might be a promising treatment strategy for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Nicole A Terpolilli
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany.,Institute for Stroke and Dementia Research, University of Munich Medical Center, Munich, Germany
| | - Sergej Feiler
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Ari Dienel
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Frank Müller
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Nicole Heumos
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Benjamin Friedrich
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - John Stover
- Department of Surgery, University of Zurich, Switzerland
| | - Serge Thal
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Karsten Schöller
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany .,Institute for Stroke and Dementia Research, University of Munich Medical Center, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
41
|
Akter F, Coghlan G, de Mel A. Nitric oxide in paediatric respiratory disorders: novel interventions to address associated vascular phenomena? Ther Adv Cardiovasc Dis 2016; 10:256-70. [PMID: 27215618 DOI: 10.1177/1753944716649893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) has a significant role in modulating the respiratory system and is being exploited therapeutically. Neonatal respiratory failure can affect around 2% of all live births and is responsible for over one third of all neonatal mortality. Current treatment method with inhaled NO (iNO) has demonstrated great benefits to patients with persistent pulmonary hypertension, bronchopulmonary dysplasia and neonatal respiratory distress syndrome. However, it is not without its drawbacks, which include the need for patients to be attached to mechanical ventilators. Notably, there is also a lack of identification of subgroups amongst abovementioned patients, and homogeneity in powered studies associated with iNO, which is one of the limitations. There are significant developments in drug delivery methods and there is a need to look at alternative or supplementary methods of NO delivery that could reduce current concerns. The addition of NO-independent activators and stimulators, or drugs such as prostaglandins to work in synergy with NO donors might be beneficial. It is of interest to consider such delivery methods within the respiratory system, where controlled release of NO can be introduced whilst minimizing the production of harmful byproducts. This article reviews current therapeutic application of iNO and the state-of-the-art technology methods for sustained delivery of NO that may be adapted and developed to address respiratory disorders. We envisage this perspective would prompt active investigation of such systems for their potential clinical benefit.
Collapse
Affiliation(s)
- Farhana Akter
- UCL Centre for Nanotechnology and Regenerative Medicine; Division of Surgery and Interventional Science, UCL, UK
| | - Gerry Coghlan
- Pulmonary Hypertension Unit, Royal Free London NHS Foundation Trust, UK
| | - Achala de Mel
- Lecturer in Regenerative Medicine, UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science, University College London, Royal Free NHS Trust Hospital, 9th Floor, Room 355, Pond Street, London NW3 2QG, UK
| |
Collapse
|
42
|
Ehlert A, Manthei G, Hesselmann V, Mathias K, Bein B, Pluta R. A Case of Hyperacute Onset of Vasospasm After Aneurysmal Subarachnoid Hemorrhage and Refractory Vasospasm Treated with Intravenous and Intraventricular Nitric Oxide: A Mini Review. World Neurosurg 2016; 91:673.e11-8. [PMID: 27109628 DOI: 10.1016/j.wneu.2016.04.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND A case of hyperacute vasospasm, indicating a poor prognosis after aneurysmal subarachnoid hemorrhage (SAH), is reported, and a review is presented of the literature addressing use of nitric oxide (NO) donors in cases of refractory vasospasm and recurrent delayed cortical ischemias (DCI). CASE DESCRIPTION A 65-year-old woman was admitted within 1 hour after aneurysmal SAH (Hunt and Hess grade III, Fisher modified by Frontera grade IV). A hyperacute vasospasm had been confirmed arteriographically, the right middle cerebral artery (MCA) aneurysm was immediately coiled and a standard antivasospastic therapy was started. Within 48 hours, the patient developed cerebral vasospasm with DCI. Because the standard therapy failed to control clinical symptoms and to address severe vasospasm, an individualized rescue treatment with NO donors was initiated. A continuous intravenous molsidomine infusion was started and clinical stabilization was achieved for a week (Hunt and Hess grade I; World Federation of Neurological Surgeons grade I; Glasgow Coma Scale score, 15) after which vasospasm and DCI recurred. During a subsequent DCI, we escalated NO donor therapy by adding intraventricular boluses of sodium nitroprusside (SNP). Over the course of the following 22 days, 7 transient DCIs (Glasgow Coma Scale score, 8) were treated with boluses of SNP during continued molsidomine therapy and each time vasospasm and DCI were completely reversed. Despite initial poor prognosis, the clinical outcome was excellent; at 3, 6, and 12 months follow-up the patient's modified National Institutes of Health-Stroke Scale and modified Rankin Scale scores were 0, with no cognitive deficits. CONCLUSIONS The review of the literature suggested that combined intravenous molsidomine with intraventricular SNP treatment reversed refractory, recurrent vasospasm and DCIs probably by addressing the hemoglobin NO sink effect, NO depletion, and decreased NO availability after aneurysmal SAH.
Collapse
Affiliation(s)
- Angelika Ehlert
- Department of Neurosurgery, Asklepios Klinik St. Georg, Hamburg, Germany.
| | - Gerd Manthei
- Department of Neurosurgery, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Volker Hesselmann
- Department of Neuroradiology, Asklepios Clinic North, Hamburg, Germany
| | - Klaus Mathias
- Department of Neuroradiology, Asklepios Clinic, St. Georg, Hamburg, Germany
| | - Berthold Bein
- Department of Anesthesiology, Asklepios Clinic, St. Georg, Hamburg, Germany
| | - Ryszard Pluta
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institute of Health, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Kayano ACAV, Dos-Santos JCK, Bastos MF, Carvalho LJ, Aliberti J, Costa FTM. Pathophysiological Mechanisms in Gaseous Therapies for Severe Malaria. Infect Immun 2016; 84:874-882. [PMID: 26831465 PMCID: PMC4807480 DOI: 10.1128/iai.01404-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over 200 million people worldwide suffer from malaria every year, a disease that causes 584,000 deaths annually. In recent years, significant improvements have been achieved on the treatment of severe malaria, with intravenous artesunate proving superior to quinine. However, mortality remains high, at 8% in children and 15% in adults in clinical trials, and even worse in the case of cerebral malaria (18% and 30%, respectively). Moreover, some individuals who do not succumb to severe malaria present long-term cognitive deficits. These observations indicate that strategies focused only on parasite killing fail to prevent neurological complications and deaths associated with severe malaria, possibly because clinical complications are associated in part with a cerebrovascular dysfunction. Consequently, different adjunctive therapies aimed at modulating malaria pathophysiological processes are currently being tested. However, none of these therapies has shown unequivocal evidence in improving patient clinical status. Recently, key studies have shown that gaseous therapies based mainly on nitric oxide (NO), carbon monoxide (CO), and hyperbaric (pressurized) oxygen (HBO) alter vascular endothelium dysfunction and modulate the host immune response to infection. Considering gaseous administration as a promising adjunctive treatment against severe malaria cases, we review here the pathophysiological mechanisms and the immunological aspects of such therapies.
Collapse
Affiliation(s)
- Ana Carolina A V Kayano
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - João Conrado K Dos-Santos
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Marcele F Bastos
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Leonardo J Carvalho
- Laboratory of Malaria Research, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Júlio Aliberti
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Fabio T M Costa
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
44
|
Lux A, Pokreisz P, Swinnen M, Caluwe E, Gillijns H, Szelid Z, Merkely B, Janssens SP. Concomitant Phosphodiesterase 5 Inhibition Enhances Myocardial Protection by Inhaled Nitric Oxide in Ischemia-Reperfusion Injury. J Pharmacol Exp Ther 2016; 356:284-92. [PMID: 26621143 DOI: 10.1124/jpet.115.227850] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/23/2015] [Indexed: 03/08/2025] Open
Abstract
Enhanced cyclic guanosine monophosphate (cGMP) signaling may attenuate myocardial ischemia-reperfusion injury (I/R) and improve left ventricular (LV) functional recovery after myocardial infarction (MI). We investigated the cardioprotection afforded by inhaled NO (iNO), the phosphodiesterase 5 (PDE5)-specific inhibitor tadalafil (TAD), or their combination (iNO+TAD) in C57Bl6J mice subjected to 6-minute left anterior descending artery ligation followed by reperfusion. We measured plasma and cardiac concentrations of cGMP during early reperfusion, quantified myocardial necrosis and inflammation by serial troponin-I (TnI) and myeloperoxidase-positive cell infiltration at day 3, and evaluated LV function and remodeling after 4 weeks using echocardiography and pressure-conductance catheterization. Administration of iNO, TAD, or both during I/R was safe and hemodynamically well tolerated. Compared with untreated mice (CON), only iNO+TAD increased plasma and cardiac-cGMP levels during early reperfusion (80 ± 12 versus 36 ± 6 pmol/ml and 0.15 ± 0.02 versus 0.05 ± 0.01 pmol/mg protein, P < 0.05 for both). Moreover, iNO+TAD reduced TnI at 4 hours to a greater extent (P < 0.001 versus CON) than either alone (P < 0.05 versus CON) and was associated with significantly less myocardial inflammatory cell infiltration at day 3. After 4 weeks and compared with CON, iNO+TAD was associated with increased fractional shortening (43 ± 1 versus 33 ± 2%, P < 0.01), larger stroke volumes (14.9 ± 1.2 versus 10.2 ± 0.9 μl, P < 0.05), enhanced septal and posterior wall thickening (P < 0.05 and P < 0.001, respectively), and attenuated LV dilatation (P < 0.001), whereas iNO or TAD alone conferred less benefit. Thus, iNO+TAD has superior efficacy to limit early reperfusion injury and attenuate adverse LV remodeling. Combination of inhaled NO with a long-acting PDE5 inhibitor may represent a promising strategy to reduce ischemic damage following reperfusion and better preserve LV function.
Collapse
Affiliation(s)
- Arpad Lux
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Peter Pokreisz
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Melissa Swinnen
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Ellen Caluwe
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Hilde Gillijns
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Zsolt Szelid
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| | - Stefan P Janssens
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary (A.L., Zs.Sz., B.M.); Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium (P.P., M.S., E.C., H.G., S.P.J.); Division of Cardiology, University Hospitals KU Leuven, Leuven, Belgium (S.P.J.)
| |
Collapse
|
45
|
Compain JD, Stanbury M, Trejo M, Chardon-Noblat S. Carbonyl-Terpyridyl-Manganese Complexes: Syntheses, Crystal Structures, and Photo-Activated Carbon Monoxide Release Properties. Eur J Inorg Chem 2015. [DOI: 10.1002/ejic.201500973] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Pulmonary Protection Strategies in Cardiac Surgery: Are We Making Any Progress? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:416235. [PMID: 26576223 PMCID: PMC4630421 DOI: 10.1155/2015/416235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/12/2015] [Indexed: 01/19/2023]
Abstract
Pulmonary dysfunction is a common complication of cardiac surgery. The mechanisms involved in the development of pulmonary dysfunction are multifactorial and can be related to the activation of inflammatory and oxidative stress pathways. Clinical manifestation varies from mild atelectasis to severe respiratory failure. Managing pulmonary dysfunction postcardiac surgery is a multistep process that starts before surgery and continues during both the operative and postoperative phases. Different pulmonary protection strategies have evolved over the years; however, the wide acceptance and clinical application of such techniques remain hindered by the poor level of evidence or the sample size of the studies. A better understanding of available modalities and/or combinations can result in the development of customised strategies for the different cohorts of patients with the potential to hence maximise patients and institutes benefits.
Collapse
|
47
|
Waltz P, Escobar D, Botero AM, Zuckerbraun BS. Nitrate/Nitrite as Critical Mediators to Limit Oxidative Injury and Inflammation. Antioxid Redox Signal 2015; 23:328-39. [PMID: 26140517 PMCID: PMC4692126 DOI: 10.1089/ars.2015.6256] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) is a critical signaling molecule marked by complex chemistry and varied biological responses depending on the context of the redox environment. In the setting of inflammation, NO can not only contribute to tissue injury and be causative of oxidative damage but can also signal as an adaptive molecule to limit inflammatory signaling in multiple cell types and tissues. RECENT ADVANCES An advance in our understanding of NO biology was the recognition of the nitrate-nitrite-NO axis, whereby nitrate (predominantly from dietary sources) could be converted to nitrite and nitrite could be reduced to NO. CRITICAL ISSUES Intriguingly, the recognition of multiple enzymes that serve as nitrite reductases in the setting of hypoxia or ischemia established the concept of nitrite as a circulating endocrine reservoir of NO, with the selective release of NO at sites that were primed for this reaction. This review highlights the anti-inflammatory roles of nitrite in numerous clinical conditions, including ischemia/reperfusion, transplant, cardiac arrest, and vascular injury, and in gastrointestinal inflammation. FUTURE DIRECTIONS These preclinical and clinical investigations set up further clinical trials and studies that elucidate the endogenous role this pathway plays in protection against inflammatory signaling.
Collapse
Affiliation(s)
- Paul Waltz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel Escobar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ana Maria Botero
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian S. Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Abstract
Advances in surgical technology have allowed for previously unconsidered therapeutic interventions. However, the complexity and invasiveness of surgical procedures are not without adverse consequences. Nitric oxide's fundamental role in a host of physiological processes, including angiogenesis, wound and bone healing, thromboresistance, smooth muscle relaxation and inflammation makes it a significant player in accelerating wound healing and mitigating the inflammation of ischemia reperfusion injury common to surgical procedures. In addition, the therapeutic properties of NO have been harnessed for the prophylactic treatment of implant infection and graft failure. In this article, we will discuss the mechanism by which NO mediates these processes, and its perioperative translational applications.
Collapse
|
49
|
Abstract
Inhaled nitric oxide (iNO) has been used extensively to treat pulmonary hypertension primarily in newborns. This therapy is a safe and effective therapy to improve the matching between airway ventilation and blood oxygenation. A key conceptual component of iNO therapy is that effects are limited to the pulmonary compartment thereby avoiding unwanted systemic effects. The mechanism underlying this model is that any NO entering the blood stream is rapidly oxidized to nitrate, a relatively inert anion that is excreted. Mediating this oxidation is oxyhemoglobin that becomes oxidized to methemoglobin, accumulation of which is limited by erythrocyte methemoglobin reductase. In this article, we discuss studies that dismiss the notion that once in the blood stream iNO is inactivated and show that a surprising result of iNO therapy is the formation of stable NO-derived products that circulate and can elicit NO-dependent signaling in extra-pulmonary tissues. This pathway has the potential to open up new applications for iNO for treatment of systemic diseases associated with loss of NO signaling.
Collapse
|
50
|
Berra L, Pinciroli R, Stowell CP, Wang L, Yu B, Fernandez BO, Feelisch M, Mietto C, Hod EA, Chipman D, Scherrer-Crosbie M, Bloch KD, Zapol WM. Autologous transfusion of stored red blood cells increases pulmonary artery pressure. Am J Respir Crit Care Med 2015; 190:800-7. [PMID: 25162920 DOI: 10.1164/rccm.201405-0850oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RATIONALE Transfusion of erythrocytes stored for prolonged periods is associated with increased mortality. Erythrocytes undergo hemolysis during storage and after transfusion. Plasma hemoglobin scavenges endogenous nitric oxide leading to systemic and pulmonary vasoconstriction. OBJECTIVES We hypothesized that transfusion of autologous blood stored for 40 days would increase the pulmonary artery pressure in volunteers with endothelial dysfunction (impaired endothelial production of nitric oxide). We also tested whether breathing nitric oxide before and during transfusion could prevent the increase of pulmonary artery pressure. METHODS Fourteen obese adults with endothelial dysfunction were enrolled in a randomized crossover study of transfusing autologous, leukoreduced blood stored for either 3 or 40 days. Volunteers were transfused with 3-day blood, 40-day blood, and 40-day blood while breathing 80 ppm nitric oxide. MEASUREMENTS AND MAIN RESULTS The age of volunteers was 41 ± 4 years (mean ± SEM), and their body mass index was 33.4 ± 1.3 kg/m(2). Plasma hemoglobin concentrations increased after transfusion with 40-day and 40-day plus nitric oxide blood but not after transfusing 3-day blood. Mean pulmonary artery pressure, estimated by transthoracic echocardiography, increased after transfusing 40-day blood (18 ± 2 to 23 ± 2 mm Hg; P < 0.05) but did not change after transfusing 3-day blood (17 ± 2 to 18 ± 2 mm Hg; P = 0.5). Breathing nitric oxide decreased pulmonary artery pressure in volunteers transfused with 40-day blood (17 ± 2 to 12 ± 1 mm Hg; P < 0.05). CONCLUSIONS Transfusion of autologous leukoreduced blood stored for 40 days was associated with increased plasma hemoglobin levels and increased pulmonary artery pressure. Breathing nitric oxide prevents the increase of pulmonary artery pressure produced by transfusing stored blood. Clinical trial registered with www.clinicaltrials.gov (NCT 01529502).
Collapse
Affiliation(s)
- Lorenzo Berra
- 1 Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|