1
|
Chin K, Jiang H, Steinberg BE, Goldenberg NM, Desjardins JF, Kabir G, Liu E, Vanama R, Baker AJ, Deschamps A, Simpson JA, Maynes JT, Vinogradov SA, Connelly KA, Mazer CD, Hare GMT. Bilateral nephrectomy impairs cardiovascular function and cerebral perfusion in a rat model of acute hemodilutional anemia. J Appl Physiol (1985) 2024; 136:1245-1259. [PMID: 38385183 DOI: 10.1152/japplphysiol.00858.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/30/2024] [Accepted: 02/15/2024] [Indexed: 02/23/2024] Open
Abstract
Anemia and renal failure are independent risk factors for perioperative stroke, prompting us to assess the combined impact of acute hemodilutional anemia and bilateral nephrectomy (2Nx) on microvascular brain Po2 (PBro2) in a rat model. Changes in PBro2 (phosphorescence quenching) and cardiac output (CO, echocardiography) were measured in different groups of anesthetized Sprague-Dawley rats (1.5% isoflurane, n = 5-8/group) randomized to Sham 2Nx or 2Nx and subsequently exposed to acute hemodilutional anemia (50% estimated blood volume exchange with 6% hydroxyethyl starch) or time-based controls (no hemodilution). Outcomes were assessed by ANOVA with significance assigned at P < 0.05. At baseline, 2Nx rats demonstrated reduced CO (49.9 ± 9.4 vs. 66.3 ± 19.3 mL/min; P = 0.014) and PBro2 (21.1 ± 2.9 vs. 32.4 ± 3.1 mmHg; P < 0.001) relative to Sham 2Nx rats. Following hemodilution, 2Nx rats demonstrated a further decrease in PBro2 (15.0 ± 6.3 mmHg, P = 0.022). Hemodiluted 2Nx rats did not demonstrate a comparable increase in CO after hemodilution compared with Sham 2Nx (74.8 ± 22.4 vs. 108.9 ± 18.8 mL/min, P = 0.003) that likely contributed to the observed reduction in PBro2. This impaired CO response was associated with reduced fractional shortening (33 ± 9 vs. 51 ± 5%) and increased left ventricular end-systolic volume (156 ± 51 vs. 72 ± 15 µL, P < 0.001) suggestive of systolic dysfunction. By contrast, hemodiluted Sham 2Nx animals demonstrated a robust increase in CO and preserved PBro2. These data support the hypothesis that the kidney plays a central role in maintaining cerebral perfusion and initiating the adaptive increase in CO required to optimize PBro2 during acute anemia.NEW & NOTEWORTHY This study has demonstrated that bilateral nephrectomy acutely impaired cardiac output (CO) and microvascular brain Po2 (PBro2), at baseline. Following acute hemodilution, nephrectomy prevented the adaptive increase in CO associated with acute hemodilution leading to a further reduction in PBro2, accentuating the degree of cerebral tissue hypoxia. These data support a role for the kidney in maintaining PBro2 and initiating the increase in CO that optimized brain perfusion during acute anemia.
Collapse
Affiliation(s)
- Kyle Chin
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Helen Jiang
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin E Steinberg
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Neil M Goldenberg
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Francois Desjardins
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Golam Kabir
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Elaine Liu
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ramesh Vanama
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada
| | - Andrew J Baker
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Alain Deschamps
- Institut de Cardiologie de Montréal, Université de Montréal, Montreal Quebec, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- IMPART investigator team Canada (https://impart.team/), Saint John, New Brunswick, Canada
| | - Jason T Maynes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Molecular Medicine, Hospital for Sick Children's Research Institute, Toronto, Ontario, Canada
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kim A Connelly
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - C David Mazer
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Gregory M T Hare
- Department of Anesthesiology and Pain Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- IMPART investigator team Canada (https://impart.team/), Saint John, New Brunswick, Canada
| |
Collapse
|
2
|
Khurshid F, Iqbal J, Ahmad FUD, Lodhi AH, Malik A, Akhtar S, Khan AA, Bux MI, Younis M. A combination of generated hydrogen sulfide and nitric oxide activity has a potentiated protectant effect against cisplatin induced nephrotoxicity. Heliyon 2024; 10:e29513. [PMID: 38655296 PMCID: PMC11036060 DOI: 10.1016/j.heliyon.2024.e29513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Aim Hydrogen sulfide and nitric oxide possess cytoprotective activity and in vivo, they are generated from exogenous sodium hydrosulfide and L-arginine respectively. Cisplatin is a major chemotherapeutic agent used to treat cancer and has a high incidence of nephrotoxicity as a side effect. The study aim was to explore the effects of NaHS and L-arginine or their combination on cisplatin induced nephrotoxicity in rats. Methods Wistar Kyoto rats were given a single intraperitoneal dose of cisplatin (5 mg/kg) followed either by NaHS (56 μmol/kg, i. p.), L-arginine (1.25 g/L in drinking water) or their combination daily for 28-days. Post-mortem plasma, urine and kidney samples were collected for biochemical assays and histopathological analysis. Results Cisplatin decreased body weights and increased urinary output, while plasma creatinine and urea levels were elevated, but sodium and potassium concentrations were diminished. The renal function parameters, blood urea nitrogen and creatinine clearance, were raised and decreased respectively. Regarding markers of reactive oxygen species, plasma total superoxide dismutase was reduced, whereas malondiadehyde was augmented.Cisplatin also diminished plasma and urinary H2S as well as plasma NO, while NaHS and L-arginine counteracted this activity on both redox-active molecules. Cisplatin cotreatment with NaHS, and/or L-arginine exhibited a reversal of all other measured parameters. Conclusion In current study, NaHS and L-arginine as monotherapy protected the rats from cisplatin-induced nephrotoxicity but the combination of both worked more effectively suggesting the augmented anti-inflammatory and antioxidative potential of test treatments when administered together.
Collapse
Affiliation(s)
- Faria Khurshid
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Javeid Iqbal
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Fiaz-Ud-Din Ahmad
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Arslan Hussain Lodhi
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Akhtar
- Department of Biochemistry, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Marvi Imam Bux
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Mohammed Younis
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| |
Collapse
|
3
|
|
4
|
Bajic Z, Sobot T, Skrbic R, Stojiljkovic MP, Ponorac N, Matavulj A, Djuric DM. Homocysteine, Vitamins B6 and Folic Acid in Experimental Models of Myocardial Infarction and Heart Failure—How Strong Is That Link? Biomolecules 2022; 12:biom12040536. [PMID: 35454125 PMCID: PMC9027107 DOI: 10.3390/biom12040536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death and the main cause of disability. In the last decade, homocysteine has been found to be a risk factor or a marker for cardiovascular diseases, including myocardial infarction (MI) and heart failure (HF). There are indications that vitamin B6 plays a significant role in the process of transsulfuration in homocysteine metabolism, specifically, in a part of the reaction in which homocysteine transfers a sulfhydryl group to serine to form α-ketobutyrate and cysteine. Therefore, an elevated homocysteine concentration (hyperhomocysteinemia) could be a consequence of vitamin B6 and/or folate deficiency. Hyperhomocysteinemia in turn could damage the endothelium and the blood vessel wall and induce worsening of atherosclerotic process, having a negative impact on the mechanisms underlying MI and HF, such as oxidative stress, inflammation, and altered function of gasotransmitters. Given the importance of the vitamin B6 in homocysteine metabolism, in this paper, we review its role in reducing oxidative stress and inflammation, influencing the functions of gasotransmitters, and improving vasodilatation and coronary flow in animal models of MI and HF.
Collapse
Affiliation(s)
- Zorislava Bajic
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Tanja Sobot
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Ranko Skrbic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Milos P. Stojiljkovic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Nenad Ponorac
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Amela Matavulj
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Dragan M. Djuric
- Faculty of Medicine, Institute of Medical Physiology “Richard Burian”, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
5
|
Li W, Lee SH, Kim SH. Carbon monoxide releasing molecule-2 suppresses stretch-activated atrial natriuretic peptide secretion by activating large-conductance calcium-activated potassium channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:125-133. [PMID: 35203062 PMCID: PMC8890946 DOI: 10.4196/kjpp.2022.26.2.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/15/2022]
Abstract
Carbon monoxide (CO) is a known gaseous bioactive substance found across a wide array of body systems. The administration of low concentrations of CO has been found to exert an anti-inflammatory, anti-apoptotic, anti-hypertensive, and vaso-dilatory effect. To date, however, it has remained unknown whether CO influences atrial natriuretic peptide (ANP) secretion. This study explores the effect of CO on ANP secretion and its associated signaling pathway using isolated beating rat atria. Atrial perfusate was collected for 10 min for use as a control, after which high atrial stretch was induced by increasing the height of the outflow catheter. Carbon monoxide releasing molecule-2 (CORM-2; 10, 50, 100 µM) and hemin (HO-1 inducer; 0.1, 1, 50 µM), but not CORM-3 (10, 50, 100 µM), decreased high stretch-induced ANP secretion. However, zinc porphyrin (HO-1 inhibitor) did not affect ANP secretion. The order of potency for the suppression of ANP secretion was found to be hemin > CORM-2 >> CORM-3. The suppression of ANP secretion by CORM-2 was attenuated by pretreatment with 5-hydroxydecanoic acid, paxilline, and 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one, but not by diltiazem, wortmannin, LY-294002, or NG-nitro-L-arginine methyl ester. Hypoxic conditions attenuated the suppressive effect of CORM-2 on ANP secretion. In sum, these results suggest that CORM-2 suppresses ANP secretion via mitochondrial KATP channels and large conductance Ca2+-activated K+ channels.
Collapse
Affiliation(s)
- Weijian Li
- Departments of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Sun Hwa Lee
- Departments of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University Jeonju 54907, Korea
| | - Suhn Hee Kim
- Departments of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University Jeonju 54907, Korea
| |
Collapse
|
6
|
Liu Y, Gao X, Xiao Q, Wang W, Zhu B. Correlation Between QTc Dispersion and Soluble Growth-stimulating Gene 2 Protein on the Early Prognosis of Acute Carbon Monoxide Poisoning Heart Disease. J Cardiovasc Pharmacol 2021; 78:572-580. [PMID: 34166304 DOI: 10.1097/fjc.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/05/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT This study aimed to explore the correlation between QTc dispersion (QTcd) and soluble growth-stimulating gene 2 protein (sST2) after heart rate correction in patients with acute carbon monoxide poisoning heart disease. Among the 150 patients, 35 cases had severe toxic heart disease. The concentrations of sST2, cardiac troponin I, and creatine kinase-MB in the severe group began to increase from admission, 24 hours, and 2 days, respectively, and their detected values were all higher than those in the nonsevere group and the normal control group. There were statistically significant differences in sST2 and QTcd between the poisoning, nonsevere, and normal control groups before the treatment. There was a statistically significant difference between the indexes of the poisoning groups at different degrees 2 and 3 days after poisoning. Receiver operating characteristic curve analysis confirmed the sensitivity and specificity of sST2 and QTcd. The correlation analysis showed that sST2 and QTcd levels were positively correlated with the incidence of severe heart disease at admission. Generally, the combined observation of sST2 and QTcd improved the prediction sensitivity and were early predictor indexes of toxic heart disease.
Collapse
Affiliation(s)
- Yongjian Liu
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | | | | | | | | |
Collapse
|
7
|
Abstract
Nitric oxide, studied to evaluate its role in cardiovascular physiology, has cardioprotective and therapeutic effects in cellular signaling, mitochondrial function, and in regulating inflammatory processes. Heme oxygenase (major role in catabolism of heme into biliverdin, carbon monoxide (CO), and iron) has similar effects as well. CO has been suggested as the molecule that is responsible for many of the above mentioned cytoprotective and therapeutic pathways as CO is a signaling molecule in the control of physiological functions. This is counterintuitive as toxic effects are related to its binding to hemoglobin. However, CO is normally produced in the body. Experimental evidence indicates that this toxic gas, CO, exerts cytoprotective properties related to cellular stress including the heart and is being assessed for its cytoprotective and cytotherapeutic properties. While survival of adult cardiomyocytes depends on oxidative phosphorylation (survival and resulting cardiac function is impaired by mitochondrial damage), mitochondrial biogenesis is modified by the heme oxygenase-1/CO system and can result in promotion of mitochondrial biogenesis by associating mitochondrial redox status to the redox-active transcription factors. It has been suggested that the heme oxygenase-1/CO system is important in differentiation of embryonic stem cells and maturation of cardiomyocytes which is thought to mitigate progression of degenerative cardiovascular diseases. Effects on other cardiac cells are being studied. Acute exposure to air pollution (and, therefore, CO) is associated with cardiovascular mortality, myocardial infarction, and heart failure, but changes in the endogenous heme oxygenase-1 system (and, thereby, CO) positively affect cardiovascular health. We will review the effect of CO on heart health and function in this article.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
8
|
Bae H, Kim T, Lim I. Carbon monoxide activates large-conductance calcium-activated potassium channels of human cardiac fibroblasts through various mechanisms. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:227-237. [PMID: 33859063 PMCID: PMC8050612 DOI: 10.4196/kjpp.2021.25.3.227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/21/2022]
Abstract
Carbon monoxide (CO) is a cardioprotectant and potential cardiovascular therapeutic agent. Human cardiac fibroblasts (HCFs) are important determinants of myocardial structure and function. Large-conductance Ca2+-activated K+ (BK) channel is a potential therapeutic target for cardiovascular disease. We investigated whether CO modulates BK channels and the signaling pathways in HCFs using whole-cell mode patch-clamp recordings. CO-releasing molecules (CORMs; CORM-2 and CORM-3) significantly increased the amplitudes of BK currents (IBK). The CO-induced stimulating effects on IBK were blocked by pre-treatment with specific nitric oxide synthase (NOS) blockers (L-NG-monomethyl arginine citrate and L-NG-nitroarginine methyl ester). 8-bromo-cyclic GMP increased IBK. KT5823 (inhibits PKG) or ODQ (inhibits soluble guanylate cyclase) blocked the CO-stimulating effect on IBK. Moreover, 8-bromo-cyclic AMP also increased IBK, and pre-treatment with KT5720 (inhibits PKA) or SQ22536 (inhibits adenylate cyclase) blocked the CO effect. Pre-treatment with N-ethylmaleimide (a thiol-alkylating reagent) also blocked the CO effect on IBK, and DL-dithiothreitol (a reducing agent) reversed the CO effect. These data suggest that CO activates IBK through NO via the NOS and through the PKG, PKA, and S-nitrosylation pathways.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
9
|
Chen YT, Liu CL, Chen CJ, Chen MH, Chen CY, Tsao PN, Chou HC, Chen PC. Association between short-term exposure to air pollution and sudden infant death syndrome. CHEMOSPHERE 2021; 271:129515. [PMID: 33450422 DOI: 10.1016/j.chemosphere.2020.129515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 06/12/2023]
Abstract
The association between air pollution and infant mortality has been inconsistently reported. A few studies have estimated short-term effects of air pollution on infants' health. This population-based case-control study aimed to examine the potential effects of air pollution on sudden infant death syndrome (SIDS) in the post-neonatal period in Taiwan during 1997-2002. Each case of infant death was matched with 20 randomly selected sex-matched controls who were born on the same day and were still alive. We obtained 24-h measurements of air pollutants and meteorological factors in each case and control with 1- to 14-day lags from 55 air-quality monitoring stations. After controlling for potential confounders, conditional logistic regression analysis was performed to estimate effects of air pollutants on SIDS (n = 398) and respiratory death (n = 121) among neonates. In single- and multi-pollutant models, we found that 100-ppb increment in carbon monoxide (Odds Ratio = 1.04-1.07) and 10-ppb increment in nitrogen dioxide (Odds Ratio = 1.20-1.35) with 1- to 14-day lags were associated with significant increase in SIDS, although a significant relationship between air pollution and respiratory death was not determined in 1- to 14-day lags. Short-term carbon monoxide and nitrogen dioxide exposure were associated with significant increase in SIDS in the post-neonatal period, with latency estimated within days before death.
Collapse
Affiliation(s)
- Yin-Ting Chen
- Division of Neonatology, Department of Pediatric, China Medical University Children's Hospital, Taiwan
| | - Chia-Lin Liu
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Chi-Jen Chen
- Institute of Epidemiology and Preventive Medicine, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Mei-Huei Chen
- Department of Pediatrics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Department of Pediatrics, National Taiwan University Hospital Yun-Lin Branch, Yunlin County, Taiwan
| | - Chien-Yi Chen
- Department of Pediatrics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; The Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Chieh Chou
- Department of Pediatrics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pau-Chung Chen
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Public Health, National Taiwan University College of Public Health, Taipei, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
10
|
Al-Owais MM, Steele DS, Holden AV, Benson AP. Deterministic and Stochastic Cellular Mechanisms Contributing to Carbon Monoxide Induced Ventricular Arrhythmias. Front Pharmacol 2021; 12:651050. [PMID: 33995065 PMCID: PMC8113948 DOI: 10.3389/fphar.2021.651050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/12/2021] [Indexed: 11/26/2022] Open
Abstract
Chronic exposure to low levels of Carbon Monoxide is associated with an increased risk of cardiac arrhythmia. Microelectrode recordings from rat and guinea pig single isolated ventricular myocytes exposed to CO releasing molecule CORM-2 and excited at 0.2/s show repolarisation changes that develop over hundreds of seconds: action potential prolongation by delayed repolarisation, EADs, multiple EADs and oscillations around the plateau, leading to irreversible repolarisation failure. The measured direct effects of CO on currents in these cells, and ion channels expressed in mammalian systems showed an increase in prolonged late Na+, and a decrease in the maximal T- and L-type Ca++. peak and late Na+, ultra-rapid delayed, delayed rectifier, and the inward rectifier K+ currents. Incorporation of these CO induced changes in maximal currents in ventricular cell models; (Gattoni et al., J. Physiol., 2016, 594, 4193-4224) (rat) and (Luo and Rudy, Circ. Res., 1994, 74, 1071-1096) (guinea-pig) and human endo-, mid-myo- and epi-cardial (O'Hara et al., PLoS Comput. Biol., 2011, 7, e1002061) models, by changes in maximal ionic conductance reproduces these repolarisation abnormalities. Simulations of cell populations with Gaussian distributions of maximal conductance parameters predict a CO induced increase in APD and its variability. Incorporation of these predicted CO induced conductance changes in human ventricular cell electrophysiology into ventricular tissue and wall models give changes in indices for the probability of the initiation of re-entrant arrhythmia.
Collapse
Affiliation(s)
- Moza M. Al-Owais
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | |
Collapse
|
11
|
Underner M, Perriot J, Peiffer G, Dewitte J, Jaafari N. Republication de : Intoxication au monoxyde de carbone chez les fumeurs actifs ou passifs de chicha. JOURNAL EUROPÉEN DES URGENCES ET DE RÉANIMATION 2021; 33:33-45. [DOI: 10.1016/j.jeurea.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
12
|
Haines DD, Tosaki A. Heme Degradation in Pathophysiology of and Countermeasures to Inflammation-Associated Disease. Int J Mol Sci 2020; 21:ijms21249698. [PMID: 33353225 PMCID: PMC7766613 DOI: 10.3390/ijms21249698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The class of tetrapyrrol "coordination complexes" called hemes are prosthetic group components of metalloproteins including hemoglobin, which provide functionality to these physiologically essential macromolecules by reversibly binding diatomic gasses, notably O2, which complexes to ferrous (reduced/Fe(II)) iron within the heme porphyrin ring of hemoglobin in a pH- and PCO2-dependent manner-thus allowing their transport and delivery to anatomic sites of their function. Here, pathologies associated with aberrant heme degradation are explored in the context of their underlying mechanisms and emerging medical countermeasures developed using heme oxygenase (HO), its major degradative enzyme and bioactive metabolites produced by HO activity. Tissue deposits of heme accumulate as a result of the removal of senescent or damaged erythrocytes from circulation by splenic macrophages, which destroy the cells and internal proteins, including hemoglobin, leaving free heme to accumulate, posing a significant toxicogenic challenge. In humans, HO uses NADPH as a reducing agent, along with molecular oxygen, to degrade heme into carbon monoxide (CO), free ferrous iron (FeII), which is sequestered by ferritin protein, and biliverdin, subsequently metabolized to bilirubin, a potent inhibitor of oxidative stress-mediated tissue damage. CO acts as a cellular messenger and augments vasodilation. Nevertheless, disease- or trauma-associated oxidative stressors sufficiently intense to overwhelm HO may trigger or exacerbate a wide range of diseases, including cardiovascular and neurologic syndromes. Here, strategies are described for counteracting the effects of aberrant heme degradation, with a particular focus on "bioflavonoids" as HO inducers, shown to cause amelioration of severe inflammatory diseases.
Collapse
Affiliation(s)
- Donald David Haines
- Advanced Biotherapeutics, London W2 1EB, UK;
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Arpad Tosaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: ; Tel./Fax: +36-52-255586
| |
Collapse
|
13
|
Calhau IB, Gomes AC, Bruno SM, Coelho AC, Magalhães CIR, Romão CC, Valente AA, Gonçalves IS, Pillinger M. One‐Pot Intercalation Strategy for the Encapsulation of a CO‐Releasing Organometallic Molecule in a Layered Double Hydroxide. Eur J Inorg Chem 2020. [DOI: 10.1002/ejic.202000202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Isabel B. Calhau
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Ana C. Gomes
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Sofia M. Bruno
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Ana C. Coelho
- Instituto de Tecnologia Química e Biológica António Xavier Universidade Nova de Lisboa Av. da República, EAN 2780‐157 Oeiras Portugal
| | - Clara I. R. Magalhães
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Carlos C. Romão
- Instituto de Tecnologia Química e Biológica António Xavier Universidade Nova de Lisboa Av. da República, EAN 2780‐157 Oeiras Portugal
| | - Anabela A. Valente
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Isabel S. Gonçalves
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Martyn Pillinger
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| |
Collapse
|
14
|
Abstract
This review is focusing on the understanding of various factors and components governing and controlling the occurrence of ventricular arrhythmias including (i) the role of various ion channel-related changes in the action potential (AP), (ii) electrocardiograms (ECGs), (iii) some important arrhythmogenic mediators of reperfusion, and pharmacological approaches to their attenuation. The transmembrane potential in myocardial cells is depending on the cellular concentrations of several ions including sodium, calcium, and potassium on both sides of the cell membrane and active or inactive stages of ion channels. The movements of Na+, K+, and Ca2+ via cell membranes produce various currents that provoke AP, determining the cardiac cycle and heart function. A specific channel has its own type of gate, and it is opening and closing under specific transmembrane voltage, ionic, or metabolic conditions. APs of sinoatrial (SA) node, atrioventricular (AV) node, and Purkinje cells determine the pacemaker activity (depolarization phase 4) of the heart, leading to the surface manifestation, registration, and evaluation of ECG waves in both animal models and humans. AP and ECG changes are key factors in arrhythmogenesis, and the analysis of these changes serve for the clarification of the mechanisms of antiarrhythmic drugs. The classification of antiarrhythmic drugs may be based on their electrophysiological properties emphasizing the connection between basic electrophysiological activities and antiarrhythmic properties. The review also summarizes some important mechanisms of ventricular arrhythmias in the ischemic/reperfused myocardium and permits an assessment of antiarrhythmic potential of drugs used for pharmacotherapy under experimental and clinical conditions.
Collapse
Affiliation(s)
- Arpad Tosaki
- Department of Pharmacology, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
15
|
Underner M, Perriot J, Peiffer G, Dewitte JD, Jaafari N. [Carbon monoxide poisoning in active or passive shisha smokers]. Rev Mal Respir 2020; 37:376-388. [PMID: 32334968 DOI: 10.1016/j.rmr.2019.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/09/2019] [Indexed: 02/05/2023]
Abstract
In France, shisha (narghile) smoking is increasingly popular among adolescents and young adults and is generally thought to be less harmful and addictive than cigarettes. This systematic review of data on carbon monoxide (CO) poisoning in active or passive shisha tobacco smokers selected 17 studies. Sixteen case reports, including 39 patients (mean age: 22.3 years; males: 51.3%), described acute carbon monoxide poisoning in active shisha smokers. The most common symptoms were dizziness, headache, and nausea. Loss of consciousness occurred in 43.6% of patients. Two patients had an epileptic seizure. The mean carboxy-haemoglobin (HbCO) blood level was 17.3%. Electrocardiographic changes were present in five patients. Most patients were treated with normobaric oxygen therapy while only four received hyperbaric oxygen therapy; two of whom were non tobacco smokers exposed to shisha smoke during their work. The outcome was favourable in all patients. Shisha use must be suspected in cases of CO poisoning, especially in adolescents and young adults. Practitioners must help shisha users to stop their consumption.
Collapse
Affiliation(s)
- M Underner
- Unité de recherche clinique, centre hospitalier Henri-Laborit, université de Poitiers, 370, avenue Jacques-Cœur, CS 10587, 86021 Poitiers, France.
| | - J Perriot
- Dispensaire Emile-Roux, CLAT 63, centre de Tabacologie, 63100 Clermont-Ferrand, France
| | - G Peiffer
- Service de pneumologie, CHR de Metz-Thionville, 57038 Metz, France
| | - J D Dewitte
- Laboratoire d'étude et de recherche en sociologie (EA 3149), Université de Brest, Bretagne-Occidentale, 29200 Brest, France; Service de Santé au travail et des maladies liées à l'environnement, CHRU de Morvan, 29609 Brest, France
| | - N Jaafari
- Unité de recherche clinique, centre hospitalier Henri-Laborit, université de Poitiers, 370, avenue Jacques-Cœur, CS 10587, 86021 Poitiers, France
| |
Collapse
|
16
|
Amorim AL, Guerreiro A, Glitz VA, Coimbra DF, Bortoluzzi AJ, Caramori GF, Braga AL, Neves A, Bernardes GJL, Peralta RA. Synthesis, characterization and photoinduced CO-release by manganese( i) complexes. NEW J CHEM 2020. [DOI: 10.1039/d0nj02260h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Three new photoCORM, two with non two with nonbonding pyridine and one with benzyl group, were synthesised, and their CO-releasing properties evaluated for with regards to their elusive binding mode.
Collapse
Affiliation(s)
- André L. Amorim
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | - Ana Guerreiro
- Instituto de Medicina Molecular
- Faculdade de Medicina
- Universidade de Lisboa
- Avenida Professor Egas Moniz
- Lisboa
| | - Vinícius A. Glitz
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | - Daniel F. Coimbra
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | | | - Giovanni F. Caramori
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | - Antonio L. Braga
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | - Ademir Neves
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| | - Gonçalo J. L. Bernardes
- Instituto de Medicina Molecular
- Faculdade de Medicina
- Universidade de Lisboa
- Avenida Professor Egas Moniz
- Lisboa
| | - Rosely A. Peralta
- Departamento de Química
- Universidade Federal de Santa Catarina
- Florianópolis
- Brazil
| |
Collapse
|
17
|
Djuric M, Nikolic Turnic T, Kostic S, Radonjic K, Jeremic J, Petkovic A, Bradic J, Milosavljevic I, Srejovic I, Zivkovic V, Djuric D, Jakovljevic V, Stevanovic P. Inhibition of gasotransmitters production and calcium influx affect cardiodynamic variables and cardiac oxidative stress in propofol-anesthetized male Wistar rats. Can J Physiol Pharmacol 2019; 97:850-856. [DOI: 10.1139/cjpp-2018-0719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been assumed that the cardioprotective effects of propofol are due to its non-anesthetic pleiotropic cardiac and vasodilator effects, in which gasotransmitters (NO, H2S, and CO) as well as calcium influx could be involved. The study on isolated rat heart was performed using 4 experimental groups (n = 7 in each): (1) bolus injection of propofol (100 mg/kg body mass, i.p.); (2) L-NAME (NO synthase inhibitor, 60 mg/kg body mass, i.p.) + propofol; (3) DL-PAG (H2S synthase inhibitor, 50 mg/kg body mass, i.p.) + propofol; (4) ZnPPIX (CO synthase inhibitor, 50 μmol/kg body mass, i.p.) + propofol. Before and after the verapamil (3 μmol/L) administration, cardiodynamic parameters were recorded (dp/dtmax, dp/dtmin, systolic left ventricular pressure, diastolic left ventricular pressure, heart rate, coronary flow), as well as coronary and cardiac oxidative stress parameters. The results showed significant increases of diastolic left ventricular pressure following NO and CO inhibition, but also increases of coronary flow following H2S and CO inhibition. Following verapamil administration, significant decreases of dp/dtmax were noted after NO and CO inhibition, then increase of diastolic left ventricular pressure following CO inhibition, and increase of coronary flow following NO, H2S, or CO inhibition. Oxidative stress markers were increased but catalase activity was significantly decreased in cardiac tissue. Gasotransmitters and calcium influx are involved in pleiotropic cardiovascular effects of propofol in male Wistar rats.
Collapse
Affiliation(s)
- M. Djuric
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, University Clinical Hospital Center “Dr. Dragisa Misovic - Dedinje”, Belgrade, Serbia
| | - T. Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - S. Kostic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - K. Radonjic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - J. Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - A. Petkovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - J. Bradic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - I. Milosavljevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - I. Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - V. Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - D. Djuric
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - V. Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical, University IM Sechenov, Moscow, Russian Federation
| | - P. Stevanovic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, University Clinical Hospital Center “Dr. Dragisa Misovic - Dedinje”, Belgrade, Serbia
| |
Collapse
|
18
|
Wysokiński D, Lewandowska P, Zątak D, Juszczak M, Kluska M, Lizińska D, Rudolf B, Woźniak K. Photoactive CO-releasing complexes containing iron - genotoxicity and ability in HO-1 gene induction in HL-60 cells. Toxicol Res (Camb) 2019; 8:544-551. [PMID: 31367337 PMCID: PMC6621133 DOI: 10.1039/c9tx00070d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
This paper presents the results of research on the biological properties of two photoactive CO-releasing molecules containing iron, i.e. (η5-C5H5)Fe(CO)2(η1-N-maleimidato) (complex A) and (η5-C5H5)Fe(CO)2(η1-N-succinimidato) (complex B). We studied their cytotoxicity, genotoxicity and the ability of inducing the HO-1 gene in HL-60 cells. We also investigated the kinetics of DNA damage repair induced by complexes A and B. We demonstrated that complex B was not toxic to HL-60 cells in high doses (above 100 μM). The ability to induce DNA damage was higher for complex A. Importantly, there was no difference in irradiated and non-irradiated cells for both complexes. DNA damage induced by complex B was repaired efficiently, while the repair of DNA damage induced by complex A was disturbed. Complex B had a minor effect on HO-1 gene expression (less than 2-fold induction), while complex A had induced HO-1 gene expression to a great extent (over 17-fold for 10 μM) - similarly in irradiated and non-irradiated HL-60 cells. The results of our research indicate that the ability of both complexes to damage DNA and to upregulate HO-1 gene expression is not related to the release of CO. Further research is needed to test whether these compounds can be considered as potential CO carriers in humans.
Collapse
Affiliation(s)
- Daniel Wysokiński
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| | - Patrycja Lewandowska
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| | - Daria Zątak
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| | - Michał Juszczak
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| | - Magdalena Kluska
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| | - Daria Lizińska
- Department of Organic Chemistry , Faculty of Chemistry , University of Lodz , 91-403 Lodz , Poland
| | - Bogna Rudolf
- Department of Organic Chemistry , Faculty of Chemistry , University of Lodz , 91-403 Lodz , Poland
| | - Katarzyna Woźniak
- Department of Molecular Genetics , Faculty of Biology and Environmental Protection , University of Lodz , 90-236 , Lodz , Poland .
| |
Collapse
|
19
|
Zhang LM, Zhang DX. The Dual Neuroprotective-Neurotoxic Effects of Sevoflurane After Hemorrhagic Shock Injury. J Surg Res 2019; 235:591-599. [DOI: 10.1016/j.jss.2018.10.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/08/2018] [Accepted: 10/25/2018] [Indexed: 12/27/2022]
|
20
|
Ward JS, De Palo A, Aucott BJ, Moir JWB, Lynam JM, Fairlamb IJS. A biotin-conjugated photo-activated CO-releasing molecule (biotinCORM): efficient CO-release from an avidin–biotinCORM protein adduct. Dalton Trans 2019; 48:16233-16241. [DOI: 10.1039/c9dt03429c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A biotinylated carbon monoxide-releasing molecule (BiotinCORM) releases CO by photoirradiation at 400 nm; an avidin–biotinCORM adduct is an effective CO-releasing molecule.
Collapse
Affiliation(s)
| | - Alice De Palo
- Department of Chemistry
- University of York
- Heslington
- UK
| | | | | | | | | |
Collapse
|
21
|
Abstract
Inhalation of high concentrations of carbon monoxide (CO) is known to lead to serious systemic complications and neuronal disturbances. However, it has been found that not only is CO produced endogenously, but also that low concentrations can bestow beneficial effects which may be of interest in biology and medicine. As translocation of CO through the human organism is difficult, small molecules known as CO-releasing molecules (CORMs) deliver controlled amounts of CO to biological systems, and these are of great interest from a medical point of view. These actions may prevent vascular dysfunction, regulate blood pressure, inhibit blood platelet aggregation or have anti-inflammatory effects. This review summarizes the functions of various CO-releasing molecules in biology and medicine.
Collapse
|
22
|
Imbrogno S, Filice M, Cerra MC, Gattuso A. NO, CO and H 2 S: What about gasotransmitters in fish and amphibian heart? Acta Physiol (Oxf) 2018; 223:e13035. [PMID: 29338122 DOI: 10.1111/apha.13035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/25/2022]
Abstract
The gasotransmitters nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2 S), long considered only toxicant, are produced in vivo during the catabolism of common biological molecules and are crucial for a large variety of physiological processes. Mounting evidence is emerging that in poikilotherm vertebrates, as in mammals, they modulate the basal performance of the heart and the response to stress challenges. In this review, we will focus on teleost fish and amphibians to highlight the evolutionary importance in vertebrates of the cardiac control elicited by NO, CO and H2 S, and the conservation of the intracellular cascades they activate. Although many gaps are still present due to discontinuous information, we will use examples obtained by studies from our and other laboratories to illustrate the complexity of the mechanisms that, by involving gasotransmitters, allow beat-to-beat, short-, medium- and long-term cardiac homoeostasis. By presenting the latest data, we will also provide a framework in which the peculiar morpho-functional arrangement of the teleost and amphibian heart can be considered as a reference tool to decipher cardiac regulatory networks which are difficult to explore using more conventional vertebrates, such as mammals.
Collapse
Affiliation(s)
- S. Imbrogno
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. Filice
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. C. Cerra
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - A. Gattuso
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| |
Collapse
|
23
|
Kapetanaki SM, Burton MJ, Basran J, Uragami C, Moody PCE, Mitcheson JS, Schmid R, Davies NW, Dorlet P, Vos MH, Storey NM, Raven E. A mechanism for CO regulation of ion channels. Nat Commun 2018; 9:907. [PMID: 29500353 PMCID: PMC5834611 DOI: 10.1038/s41467-018-03291-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/31/2018] [Indexed: 01/08/2023] Open
Abstract
Despite being highly toxic, carbon monoxide (CO) is also an essential intracellular signalling molecule. The mechanisms of CO-dependent cell signalling are poorly defined, but are likely to involve interactions with heme proteins. One such role for CO is in ion channel regulation. Here, we examine the interaction of CO with KATP channels. We find that CO activates KATP channels and that heme binding to a CXXHX16H motif on the SUR2A receptor is required for the CO-dependent increase in channel activity. Spectroscopic and kinetic data were used to quantify the interaction of CO with the ferrous heme-SUR2A complex. The results are significant because they directly connect CO-dependent regulation to a heme-binding event on the channel. We use this information to present molecular-level insight into the dynamic processes that control the interactions of CO with a heme-regulated channel protein, and we present a structural framework for understanding the complex interplay between heme and CO in ion channel regulation.
Collapse
Affiliation(s)
- Sofia M Kapetanaki
- Department of Chemistry and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, England
| | - Mark J Burton
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - Jaswir Basran
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - Chiasa Uragami
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Peter C E Moody
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - John S Mitcheson
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - Ralf Schmid
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - Noel W Davies
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England
| | - Pierre Dorlet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Marten H Vos
- LOB, Ecole Polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau Cedex, France
| | - Nina M Storey
- Department of Molecular and Cell Biology and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 9HN, England.
| | - Emma Raven
- Department of Chemistry and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, England.
| |
Collapse
|
24
|
Cheraki M, Al-Mogren MM, Chambaud G, Francisco JS, Hochlaf M. Identification of Key Intermediates during the NO and H2S Crosstalk Signaling Pathways. J Phys Chem A 2018; 122:2877-2883. [DOI: 10.1021/acs.jpca.7b11821] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mohamed Cheraki
- Laboratoire Modélisation et Simulation Multi Echelle, Université Paris-Est, MSME UMR 8208 CNRS, 5 Boulevard Descartes, 77454 Marne-la-Vallée, France
| | - Muneerah Mogren Al-Mogren
- Chemistry Department, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Kingdom of Saudi Arabia
| | - Gilberte Chambaud
- Laboratoire Modélisation et Simulation Multi Echelle, Université Paris-Est, MSME UMR 8208 CNRS, 5 Boulevard Descartes, 77454 Marne-la-Vallée, France
| | - Joseph S. Francisco
- Department of Chemistry, University of Nebraska—Lincoln, 433 Hamilton Hall, Lincoln, Nebraska 68588-0304, United States
| | - Majdi Hochlaf
- Laboratoire Modélisation et Simulation Multi Echelle, Université Paris-Est, MSME UMR 8208 CNRS, 5 Boulevard Descartes, 77454 Marne-la-Vallée, France
| |
Collapse
|
25
|
Garg J, Krishnamoorthy P, Palaniswamy C, Khera S, Ahmad H, Jain D, Aronow WS, Frishman WH. Cardiovascular Abnormalities in Carbon Monoxide Poisoning. Am J Ther 2018; 25:e339-e348. [PMID: 24518173 DOI: 10.1097/mjt.0000000000000016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acute carbon monoxide (CO) poisoning is the most common cause of poisoning and poisoning-related death in the United States. It manifests as broad spectrum of symptoms ranging from mild headache, nausea, and fatigue to dizziness, syncope, coma, seizures resulting in cardiovascular collapse, respiratory failure, and death. Cardiovascular complications of CO poisoning has been well reported and include myocardial stunning, left ventricular dysfunction, pulmonary edema, and arrhythmias. Acute myocardial ischemia has also been reported from increased thrombogenicity due to CO poisoning. Myocardial toxicity from CO exposure is associated with increased short-term and long-term mortality. Carboxyhemoglobin (COHb) levels do not correlate well with the clinical severity of CO poisoning. Supplemental oxygen remains the cornerstone of therapy for CO poisoning. Hyperbaric oxygen therapy increases CO elimination and has been used with wide variability in patients with evidence of neurological and myocardial injury from CO poisoning, but its benefit in limiting or reversing cardiac injury is unknown. We present a comprehensive review of literature on cardiovascular manifestations of CO poisoning and propose a diagnostic algorithm for managing patients with CO poisoning.
Collapse
Affiliation(s)
- Jalaj Garg
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Parasuram Krishnamoorthy
- Department of Internal Medicine, Mount Sinai Englewood Hospital and Medical Center Englewood, NJ
| | - Chandrasekar Palaniswamy
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Sahil Khera
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Hasan Ahmad
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Diwakar Jain
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - William H Frishman
- Division of Cardiology, Department of Internal Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| |
Collapse
|
26
|
Al-Owais MM, Hettiarachchi NT, Boyle JP, Scragg JL, Elies J, Dallas ML, Lippiat JD, Steele DS, Peers C. Multiple mechanisms mediating carbon monoxide inhibition of the voltage-gated K + channel Kv1.5. Cell Death Dis 2017; 8:e3163. [PMID: 29095440 PMCID: PMC5775415 DOI: 10.1038/cddis.2017.568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Mark L Dallas
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jon D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
27
|
Al-Owais MM, Hettiarachchi NT, Kirton HM, Hardy ME, Boyle JP, Scragg JL, Steele DS, Peers C. A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K + channels in carbon monoxide-induced proarrhythmic early afterdepolarizations. FASEB J 2017; 31:4845-4854. [PMID: 28743763 PMCID: PMC5636698 DOI: 10.1096/fj.201700259r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022]
Abstract
Exposure to CO causes early afterdepolarization arrhythmias. Previous studies in rats have indicated that arrhythmias arose as a result of augmentation of the late Na+ current. The purpose of the present study was to examine the basis for CO-induced arrhythmias in guinea pig myocytes in which action potentials (APs) more closely resemble those of human myocytes. Whole-cell current- and voltage-clamp recordings were made from isolated guinea pig myocytes as well as from human embryonic kidney 293 (HEK293) cells that express wild-type or a C723S mutant form of ether-a-go-go-related gene (ERG; Kv11.1). We also monitored the formation of peroxynitrite (ONOO-) in HEK293 cells fluorimetrically. CO-applied as the CO-releasing molecule, CORM-2-prolonged the APs and induced early afterdepolarizations in guinea pig myocytes. In HEK293 cells, CO inhibited wild-type, but not C723S mutant, Kv11.1 K+ currents. Inhibition was prevented by an antioxidant, mitochondrial inhibitors, or inhibition of NO formation. CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. Our data suggest that CO induces arrhythmias in guinea pig cardiac myocytes via the ONOO--mediated inhibition of Kv11.1 K+ channels.-Al-Owais, M. M., Hettiarachchi, N. T., Kirton, H. M., Hardy, M. E., Boyle, J. P., Scragg, J. L., Steele, D. S., Peers, C. A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K+ channels in carbon monoxide-induced proarrhythmic early afterdepolarizations.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Hannah M Kirton
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Matthew E Hardy
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Derek S Steele
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| |
Collapse
|
28
|
Ahamed J, Laurence J. Role of Platelet-Derived Transforming Growth Factor-β1 and Reactive Oxygen Species in Radiation-Induced Organ Fibrosis. Antioxid Redox Signal 2017; 27:977-988. [PMID: 28562065 PMCID: PMC5649128 DOI: 10.1089/ars.2017.7064] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE This review evaluates the role of platelet-derived transforming growth factor (TGF)-β1 in oxidative stress-linked pathologic fibrosis, with an emphasis on the heart and kidney, by using ionizing radiation as a clinically relevant stimulus. Current radiation-induced organ fibrosis interventions focus on pan-neutralization of TGF-β or the use of anti-oxidants and anti-proliferative agents, with limited clinical efficacy. Recent Advances: Pathologic fibrosis represents excessive accumulation of collagen and other extracellular matrix (ECM) components after dysregulation of a balance between ECM synthesis and degradation. Targets based on endogenous carbon monoxide (CO) pathways and the use of redox modulators such as N-acetylcysteine present promising alternatives to current therapeutic regimens. CRITICAL ISSUES Ionizing radiation leads to direct DNA damage and generation of reactive oxygen species (ROS), with TGF-β1 activation via ROS, thrombin generation, platelet activation, and pro-inflammatory signaling promoting myofibroblast accumulation and ECM production. Feed-forward loops, as TGF-β1 promotes ROS, amplify these profibrotic signals, and persistent low-grade inflammation insures their perpetuation. We highlight differential roles for platelet- versus monocyte-derived TGF-β1, establishing links between canonical and noncanonical TGF-β1 signaling pathways in relationship to macrophage polarization and autophagy, and define points where pharmacologic agents can intervene. FUTURE DIRECTIONS Additional studies are needed to understand mechanisms underlying the anti-fibrotic effects of current and proposed therapeutics, based on limiting platelet TGF-β1 activity, promotion of macrophage polarization, and facilitation of collagen autophagy. Models incorporating endogenous CO and selective TGF-β1 pathways that impact the initiation and progression of pathologic fibrosis, including nuclear factor erythroid 2-related factor (Nrf2) and redox, are of particular interest. Antioxid. Redox Signal. 27, 977-988.
Collapse
Affiliation(s)
- Jasimuddin Ahamed
- 1 Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation , Oklahoma City, Oklahoma
| | - Jeffrey Laurence
- 2 Division of Hematology and Medical Oncology, Weill Cornell Medical College , New York, New York
| |
Collapse
|
29
|
HIV protease inhibitor-induced cardiac dysfunction and fibrosis is mediated by platelet-derived TGF-β1 and can be suppressed by exogenous carbon monoxide. PLoS One 2017; 12:e0187185. [PMID: 29088262 PMCID: PMC5663426 DOI: 10.1371/journal.pone.0187185] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection is an independent risk factor for cardiovascular disease. This risk is magnified by certain antiretrovirals, particularly the protease inhibitor ritonavir, but the pathophysiology of this connection is unknown. We postulated that a major mechanism for antiretroviral-associated cardiac disease is pathologic fibrosis linked to platelet activation with release and activation of transforming growth factor (TGF)-β1, and that these changes could be modeled in a murine system. We also sought to intervene utilizing inhaled carbon monoxide (CO) as proof-of-concept for therapeutics capable of regulating TGF-β1 signaling and collagen autophagy. We demonstrate decreased cardiac function indices, including cardiac output, ejection fraction and stroke volume, and prominent cardiac fibrosis, in mice exposed to pharmacological doses of ritonavir. Cardiac output and fibrosis correlated with plasma TGF-β1 levels. Mice with targeted deletion of TGF-β1 in megakaryocytes/platelets (PF4CreTgfb1flox/flox) were partially protected from ritonavir-induced cardiac dysfunction and fibrosis. Inhalation of low dose CO (250ppm), used as a surrogate for upregulation of inducible heme oxygenase/endogenous CO pathways, suppressed ritonavir-induced cardiac fibrosis. This occurred in association with modulation of canonical (Smad2) and non-canonical (p38) TGF-β1 signaling pathways. In addition, CO treatment suppressed the M1 pro-inflammatory subset of macrophages and increased M2c regulatory cells in the hearts of RTV-exposed animals. The effects of CO were dependent upon autophagy as CO did not mitigate ritonavir-induced fibrosis in autophagy-deficient LC3-/- mice. These results suggest that platelet-derived TGF-β1 contributes to ritonavir-associated cardiac dysfunction and fibrosis, extending the relevance of our findings to other antiretrovirals that also activate platelets. The anti-fibrotic effects of CO are linked to alterations in TGF-β1 signaling and autophagy, suggesting a proof-of-concept for novel interventions in HIV/antiretroviral therapy-mediated cardiovascular disease.
Collapse
|
30
|
Pinto MN, Chakraborty I, Sandoval C, Mascharak PK. Eradication of HT-29 colorectal adenocarcinoma cells by controlled photorelease of CO from a CO-releasing polymer (photoCORP-1) triggered by visible light through an optical fiber-based device. J Control Release 2017; 264:192-202. [PMID: 28866022 DOI: 10.1016/j.jconrel.2017.08.039] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/06/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
The gaseous signaling molecule carbon monoxide (CO) has recently been recognized for its wide range of physiological activity as well as its antineoplastic properties. However, site-specific delivery of this noxious gas presents a major challenge in hospital settings. In this work, a visible light-sensitive CO-releasing molecule (photoCORM) derived from manganese(I) and 2-(quinolyl)benzothiazole (qbt) namely, [Mn(CO)3(qbt)(4-vpy)](CF3SO3) (1), has been co-polymerized within a gas-permeable HEMA/EGDMA hydrogel. The resulting photoactive CO-releasing polymer (photoCORP-1) incorporates 1 such that neither the carbonyl complex nor its photoproduct(s) exits the polymer at any time. The material can be triggered to photorelease CO remotely by low-power broadband visible light (<1mWcm-2) with the aid of fiber optics technology. The CO photorelease rates of photoCORP-1 (determined by spectrophotometry) can be modulated by both the concentration of 1 in the hydrogel and the intensity of the light. A CO-delivery device has been assembled to deliver CO to a suspension of human colorectal adenocarcinoma cells (HT-29) under the control of visible light and the extent of CO-induced apoptotic death of the cancer cells has been determined via Annexin V/Propidium iodide stain and flow cytometry. This photoactive CO-releasing polymer could find use in delivering controlled doses of CO to cellular targets such as malignant tissues in remote parts of the body.
Collapse
Affiliation(s)
- Miguel N Pinto
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | - Indranil Chakraborty
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | - Cosme Sandoval
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | - Pradip K Mascharak
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA.
| |
Collapse
|
31
|
Abstract
Part I of this review discussed the similarities between embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1 [HIF-1]), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, and it focused on the common characteristics that allow cells and organisms to survive in these states. Part II of this review describes techniques by which researchers gain insight into subcellular energetics and identify potential future tools for clinicians. In particular, P nuclear magnetic resonance to measure high-energy phosphates, serum lactate measurements, the use of near-infrared spectroscopy to measure the oxidation state of cytochrome aa3, and the ability of the protoporphyrin IX-triplet state lifetime technique to measure mitochondrial oxygen tension are discussed. In addition, this review discusses novel treatment strategies such as hyperbaric oxygen, preconditioning, exercise training, therapeutic gases, as well as inhibitors of HIF-1, HIF prolyl hydroxylase, and peroxisome proliferator-activated receptors.
Collapse
Affiliation(s)
- Robert H Thiele
- From the Department of Anesthesiology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
32
|
Otterbein LE, Foresti R, Motterlini R. Heme Oxygenase-1 and Carbon Monoxide in the Heart: The Balancing Act Between Danger Signaling and Pro-Survival. Circ Res 2017; 118:1940-1959. [PMID: 27283533 DOI: 10.1161/circresaha.116.306588] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Understanding the processes governing the ability of the heart to repair and regenerate after injury is crucial for developing translational medical solutions. New avenues of exploration include cardiac cell therapy and cellular reprogramming targeting cell death and regeneration. An attractive possibility is the exploitation of cytoprotective genes that exist solely for self-preservation processes and serve to promote and support cell survival. Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. The remarkable cardioprotective effects ascribed to heme oxygenase-1 are best evidenced by its ability to regulate inflammatory processes, cellular signaling, and mitochondrial function ultimately mitigating myocardial tissue injury and the progression of vascular-proliferative disease. We discuss here new insights into the role of heme oxygenase-1 and heme on cardiovascular health, and importantly, how they might be leveraged to promote heart repair after injury.
Collapse
Affiliation(s)
- Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Roberta Foresti
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| | - Roberto Motterlini
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| |
Collapse
|
33
|
Kehr G, Erker G. Frustrated Lewis Pair Chemistry: Searching for New Reactions. CHEM REC 2017; 17:803-815. [DOI: 10.1002/tcr.201700010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Gerald Kehr
- Organisch-Chemisches Institut; Westfälische Wilhelms-Universität; 48149 Münster Corrensstr. 40 Germany
| | - Gerhard Erker
- Organisch-Chemisches Institut; Westfälische Wilhelms-Universität; 48149 Münster Corrensstr. 40 Germany
| |
Collapse
|
34
|
Cebová M, Košútová M, Pecháňová O. Cardiovascular effects of gasotransmitter donors. Physiol Res 2017; 65:S291-S307. [PMID: 27775418 DOI: 10.33549/physiolres.933441] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gasotransmitters represent a subfamily of the endogenous gaseous signaling molecules that include nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H(2)S). These particular gases share many common features in their production and function, but they fulfill their physiological tasks in unique ways that differ from those of classical signaling molecules found in tissues and organs. These gasotransmitters may antagonize or potentiate each other's cellular effects at the level of their production, their downstream molecular targets and their direct interactions. All three gasotransmitters induce vasodilatation, inhibit apoptosis directly or by increasing the expression of anti-apoptotic genes, and activate antioxidants while inhibiting inflammatory actions. NO and CO may concomitantly participate in vasorelaxation, anti-inflammation and angiogenesis. NO and H(2)S collaborate in the regulation of vascular tone. Finally, H(2)S may upregulate the heme oxygenase/carbon monoxide (HO/CO) pathway during hypoxic conditions. All three gasotransmitters are produced by specific enzymes in different cell types that include cardiomyocytes, endothelial cells and smooth muscle cells. As translational research on gasotransmitters has exploded over the past years, drugs that alter the production/levels of the gasotransmitters themselves or modulate their signaling pathways are now being developed. This review is focused on the cardiovascular effects of NO, CO, and H(2)S. Moreover, their donors as drug targeting the cardiovascular system are briefly described.
Collapse
Affiliation(s)
- M Cebová
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | |
Collapse
|
35
|
Hochlaf M. Advances in spectroscopy and dynamics of small and medium sized molecules and clusters. Phys Chem Chem Phys 2017; 19:21236-21261. [DOI: 10.1039/c7cp01980g] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Investigations of the spectroscopy and dynamics of small- and medium-sized molecules and clusters represent a hot topic in atmospheric chemistry, biology, physics, atto- and femto-chemistry and astrophysics.
Collapse
Affiliation(s)
- Majdi Hochlaf
- Université Paris-Est
- Laboratoire Modélisation et Simulation Multi Echelle
- MSME UMR 8208 CNRS
- 77454 Marne-la-Vallée
- France
| |
Collapse
|
36
|
Protective effect of erythropoietin on myocardial apoptosis in rats exposed to carbon monoxide. Life Sci 2016; 148:118-24. [PMID: 26855001 DOI: 10.1016/j.lfs.2016.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 11/21/2022]
Abstract
AIMS Cardiac complications are common in carbon monoxide (CO) poisoning and associated with high morbidity and mortality. We have previously shown that erythropoietin (EPO) could reduce CO-induced cardiac ischemia in rat. In the current study, the anti-apoptotic effect of EPO during CO cardiotoxicity was investigated in order to elucidate the mechanism of EPO anti-ischemic action. MAIN METHODS Wistar rats were exposed to CO (250, 1000 and 3000ppm). EPO (5000IU/kg) was administered to all groups by intraperitoneal injection at the end of CO exposure period. TUNEL and caspase-3 activity levels were assessed to investigate the effects of CO exposure and subsequent EPO administration on myocardial apoptosis. The changes of mitochondrial membrane potential (MMP) were also assessed with sensitive lipophilic dye JC-1 by flow cytometry. The roles of Bcl2 and Bax in EPO protective effect were investigated by Western blotting. KEY FINDINGS Myocardial apoptosis was observed following CO exposure. Moreover, mitochondrial membrane depolarization and significant reduction in Bcl2/Bax ratio were shown following CO poisoning especially at 3000ppm. On the other hand, EPO administration could effectively suppress apoptosis in myocardial cells. Also, EPO significantly prevented the CO-induced depolarization of MMP (p<0.001) and preserved Bcl2/Bax ratio (p<0.01). SIGNIFICANCE EPO reduces myocardial injury due to CO intoxication. Thus EPO could be suggested as a possible candidate for the management of CO cardiotoxicity with clinical applications.
Collapse
|
37
|
Jiménez-Pulido SB, Illán-Cabeza NA, Hueso-Ureña F, Maldonado CR, Sánchez-Sánchez P, Fernández-Liencres MP, Fernández-Gómez M, Moreno-Carretero MN. A combined experimental and DFT investigation on the structure and CO-releasing properties of mono and binuclear fac-ReI(CO)3 complexes with 5-pyridin-2-ylmethylene-amino uracils. Dalton Trans 2016; 45:15142-15154. [DOI: 10.1039/c6dt02208a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New tricarbonyl rhenium(i) complexes with 5-substituted-6-amino-1,3-dimethyluracils are reported.
Collapse
Affiliation(s)
- Sonia B. Jiménez-Pulido
- Department of Inorganic and Organic Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| | - Nuria A. Illán-Cabeza
- Department of Inorganic and Organic Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| | - Francisco Hueso-Ureña
- Department of Inorganic and Organic Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| | - Carmen R. Maldonado
- Department of Inorganic Chemistry
- Campus Fuentenueva
- University of Granada
- 18071-Granada
- Spain
| | | | - M. Paz Fernández-Liencres
- Department of Physical and Analytical Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| | - Manuel Fernández-Gómez
- Department of Physical and Analytical Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| | - Miguel N. Moreno-Carretero
- Department of Inorganic and Organic Chemistry
- Campus Las Lagunillas (B3)
- University of Jaén
- 23071-Jaén
- Spain
| |
Collapse
|
38
|
An emerging role for gasotransmitters in the control of breathing and ionic regulation in fish. J Comp Physiol B 2015; 186:145-59. [DOI: 10.1007/s00360-015-0949-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/04/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
|
39
|
Wang LW, He EYJ, Ghosh D, Day RO, Jones GRD, Subbiah RN, Holloway CJ. Severe carbon monoxide poisoning from waterpipe smoking: a public health concern. Med J Aust 2015; 202:446-7. [DOI: 10.5694/mja14.01264] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 11/11/2014] [Indexed: 11/17/2022]
Affiliation(s)
- Louis W Wang
- St Vincent's Hospital, Sydney, NSW
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW
- Victor Chang Cardiac Research Institute, Sydney, NSW
| | - Emily YJ He
- Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW
| | | | - Richard O Day
- St Vincent's Hospital, Sydney, NSW
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW
| | - Graham RD Jones
- St Vincent's Hospital, Sydney, NSW
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW
| | - Rajesh N Subbiah
- St Vincent's Hospital, Sydney, NSW
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW
- Victor Chang Cardiac Research Institute, Sydney, NSW
| | - Cameron J Holloway
- St Vincent's Hospital, Sydney, NSW
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW
- Victor Chang Cardiac Research Institute, Sydney, NSW
| |
Collapse
|
40
|
Gasomediators (·NO, CO, and H2S) and their role in hemostasis and thrombosis. Clin Chim Acta 2015; 445:115-21. [DOI: 10.1016/j.cca.2015.03.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/16/2023]
|
41
|
Elies J, Johnson E, Boyle JP, Scragg JL, Peers C. H2S does not regulate proliferation via T-type Ca2+ channels. Biochem Biophys Res Commun 2015; 461:659-64. [PMID: 25918023 DOI: 10.1016/j.bbrc.2015.04.087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/17/2015] [Indexed: 01/08/2023]
Abstract
T-type Ca(2+) channels (Cav3.1, 3.2 and 3.3) strongly influence proliferation of various cell types, including vascular smooth muscle cells (VSMCs) and certain cancers. We have recently shown that the gasotransmitter carbon monoxide (CO) inhibits T-type Ca(2+) channels and, in so doing, attenuates proliferation of VSMC. We have also shown that the T-type Ca(2+) channel Cav3.2 is selectively inhibited by hydrogen sulfide (H2S) whilst the other channel isoforms (Cav3.1 and Cav3.3) are unaffected. Here, we explored whether inhibition of Cav3.2 by H2S could account for the anti-proliferative effects of this gasotransmitter. H2S suppressed proliferation in HEK293 cells expressing Cav3.2, as predicted by our previous observations. However, H2S was similarly effective in suppressing proliferation in wild type (non-transfected) HEK293 cells and those expressing the H2S insensitive channel, Cav3.1. Further studies demonstrated that T-type Ca(2+) channels in the smooth muscle cell line A7r5 and in human coronary VSMCs strongly influenced proliferation. In both cell types, H2S caused a concentration-dependent inhibition of proliferation, yet by far the dominant T-type Ca(2+) channel isoform was the H2S-insensitive channel, Cav3.1. Our data indicate that inhibition of T-type Ca(2+) channel-mediated proliferation by H2S is independent of the channels' sensitivity to H2S.
Collapse
Affiliation(s)
- Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | - Emily Johnson
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
42
|
Peers C, Boyle JP, Scragg JL, Dallas ML, Al-Owais MM, Hettiarachichi NT, Elies J, Johnson E, Gamper N, Steele DS. Diverse mechanisms underlying the regulation of ion channels by carbon monoxide. Br J Pharmacol 2015; 172:1546-56. [PMID: 24818840 PMCID: PMC4369263 DOI: 10.1111/bph.12760] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/14/2014] [Accepted: 04/21/2014] [Indexed: 12/19/2022] Open
Abstract
Carbon monoxide (CO) is firmly established as an important, physiological signalling molecule as well as a potent toxin. Through its ability to bind metal-containing proteins, it is known to interfere with a number of intracellular signalling pathways, and such actions can account for its physiological and pathological effects. In particular, CO can modulate the intracellular production of reactive oxygen species, NO and cGMP levels, as well as regulate MAPK signalling. In this review, we consider ion channels as more recently discovered effectors of CO signalling. CO is now known to regulate a growing number of different ion channel types, and detailed studies of the underlying mechanisms of action are revealing unexpected findings. For example, there are clear areas of contention surrounding its ability to increase the activity of high conductance, Ca(2+) -sensitive K(+) channels. More recent studies have revealed the ability of CO to inhibit T-type Ca(2+) channels and have unveiled a novel signalling pathway underlying tonic regulation of this channel. It is clear that the investigation of ion channels as effectors of CO signalling is in its infancy, and much more work is required to fully understand both the physiological and the toxic actions of this gas. Only then can its emerging use as a therapeutic tool be fully and safely exploited.
Collapse
Affiliation(s)
- C Peers
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - J P Boyle
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - J L Scragg
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - M L Dallas
- School of Pharmacy, University of ReadingReading, UK
| | - M M Al-Owais
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - N T Hettiarachichi
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - J Elies
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - E Johnson
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of LeedsLeeds, UK
| | - N Gamper
- Faculty of Biological Sciences, University of LeedsLeeds, UK
| | - D S Steele
- Faculty of Biological Sciences, University of LeedsLeeds, UK
| |
Collapse
|
43
|
Abstract
SIGNIFICANCE Oxygen plays a key role in cellular metabolism and function. Oxygen delivery to cells is crucial, and a lack of oxygen such as that which occurs during myocardial infarction can be lethal. Cells should, therefore, be able to respond to changes in oxygen tension. RECENT ADVANCES Since the first studies examining the acute cellular effect of hypoxia on activation of transmitter release from glomus or type I chemoreceptor cells, it is now known that virtually all cells are able to respond to changes in oxygen tension. CRITICAL ISSUES Despite advances made in characterizing hypoxic responses, the identity of the "oxygen sensor" remains debated. Recently, more evidence has evolved as to how cardiac myocytes sense acute changes in oxygen. This review will examine the available evidence in support of acute oxygen-sensing mechanisms providing a brief historical perspective and then more detailed insights into the heart and the role of cardiac ion channels in hypoxic responses. FUTURE DIRECTIONS A further understanding of these cellular processes should result in interventions that assist in preventing the deleterious effects of acute changes in oxygen tension such as alterations in contractile function and cardiac arrhythmia.
Collapse
Affiliation(s)
- Livia C Hool
- School of Anatomy, Physiology, and Human Biology, The University of Western Australia , Crawley, Australia
| |
Collapse
|
44
|
Gamper N, Ooi L. Redox and nitric oxide-mediated regulation of sensory neuron ion channel function. Antioxid Redox Signal 2015; 22:486-504. [PMID: 24735331 PMCID: PMC4323017 DOI: 10.1089/ars.2014.5884] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Reactive oxygen and nitrogen species (ROS and RNS, respectively) can intimately control neuronal excitability and synaptic strength by regulating the function of many ion channels. In peripheral sensory neurons, such regulation contributes towards the control of somatosensory processing; therefore, understanding the mechanisms of such regulation is necessary for the development of new therapeutic strategies and for the treatment of sensory dysfunctions, such as chronic pain. RECENT ADVANCES Tremendous progress in deciphering nitric oxide (NO) and ROS signaling in the nervous system has been made in recent decades. This includes the recognition of these molecules as important second messengers and the elucidation of their metabolic pathways and cellular targets. Mounting evidence suggests that these targets include many ion channels which can be directly or indirectly modulated by ROS and NO. However, the mechanisms specific to sensory neurons are still poorly understood. This review will therefore summarize recent findings that highlight the complex nature of the signaling pathways involved in redox/NO regulation of sensory neuron ion channels and excitability; references to redox mechanisms described in other neuron types will be made where necessary. CRITICAL ISSUES The complexity and interplay within the redox, NO, and other gasotransmitter modulation of protein function are still largely unresolved. Issues of specificity and intracellular localization of these signaling cascades will also be addressed. FUTURE DIRECTIONS Since our understanding of ROS and RNS signaling in sensory neurons is limited, there is a multitude of future directions; one of the most important issues for further study is the establishment of the exact roles that these signaling pathways play in pain processing and the translation of this understanding into new therapeutics.
Collapse
Affiliation(s)
- Nikita Gamper
- 1 Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds , Leeds, United Kingdom
| | | |
Collapse
|
45
|
Peers C, Boyle JP. Oxidative modulation of K+ channels in the central nervous system in neurodegenerative diseases and aging. Antioxid Redox Signal 2015; 22:505-21. [PMID: 25333910 DOI: 10.1089/ars.2014.6007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Oxidative stress and damage are well-established components of neurodegenerative diseases, contributing to neuronal death during disease progression. Here, we consider key K(+) channels as target proteins that can undergo oxidative modulation, describe what is understood about how this influences disease progression, and consider regulation of these channels by gasotransmitters as a means of cellular protection. RECENT ADVANCES Oxidative regulation of the delayed rectifier Kv2.1 and the Ca(2+)- and voltage-sensitive BK channel are established, but recent studies contest how their redox sensitivity contributes to altered excitability, progression of neurodegenerative diseases, and healthy aging. CRITICAL ISSUES Both Kv2.1 and BK channels have recently been established as target proteins for regulation by the gasotransmitters carbon monoxide and hydrogen sulfide. Establishing the molecular basis of such regulation, and exactly how this influences excitability and vulnerability to apoptotic cell death will determine whether such regulation can be exploited for therapeutic benefit. FUTURE DIRECTIONS Developing a more comprehensive picture of the oxidative modulation of K(+) channels (and, indeed, other ion channels) within the central nervous system in health and disease will enable us to better understand processes associated with healthy aging as well as distinct processes underlying progression of neurodegenerative diseases. Advances in the growing understanding of how gasotransmitters can regulate ion channels, including redox-sensitive K(+) channels, are a research priority for this field, and will establish their usefulness in design of future approaches for the treatment of such diseases.
Collapse
Affiliation(s)
- Chris Peers
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), Faculty of Medicine and Health, University of Leeds , Leeds, United Kingdom
| | | |
Collapse
|
46
|
Carbon monoxide modulates electrical activity of murine myocardium via cGMP-dependent mechanisms. J Physiol Biochem 2015; 71:107-19. [PMID: 25670496 DOI: 10.1007/s13105-015-0387-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/25/2015] [Indexed: 10/24/2022]
Abstract
Carbon monoxide (CO) is critical in cell signaling, and inhalation of gaseous CO can impact cardiovascular physiology. We have investigated electrophysiological effects of CO and their potential cGMP-dependent mechanism in isolated preparations of murine myocardium. The standard microelectrode technique was used to record myocardial action potentials (APs). Exogenous CO (0.96 × 10(-4)-4.8 × 10(-4) M) decreased AP duration in atrial and ventricular tissue and accelerated pacemaking activity in sinoatrial node. Inhibitors of heme oxygenases (zinc and tin protoporphyrin IX), which are responsible for endogenous CO production, induced the opposite effects. Inhibitor of soluble guanylate cyclase (sGC), ODQ (10(-5) M) halved CO-induced AP shortening, while sGC activator azosidnone (10(-5) M-3 × 10(-4) M) and cGMP analog BrcGMP (3 × 10(-4) M) induced the same effects as CO. To see if CO effects are attributed to differential regulation of phosphodiesterase 2 (PDE2) and 3 (PDE3), we used inhibitors of these enzymes. Milrinone (2 × 10(-6) M), selective inhibitor of cGMP-downregulated PDE3, blocked CO-induced rhythm acceleration. EHNA(2 × 10(-6) M), which inhibits cGMP-upregulated PDE2, attenuated CO-induced AP shortening, but failed to induce any positive chronotropic effect. Our findings indicate that PDE2 activity prevails in working myocardium, while PDE3 is more active in sinoatrial node. The results suggest that cardiac effects of CO are at least partly attributed to activation of sGC and subsequent elevation of cGMP intracellular content. In sinoatrial node, this leads to PDE3 inhibition, increased cAMP content, and positive chronotropy, while it also causes PDE2 stimulation in working myocardium, thereby enhancing cAMP degradation and producing AP shortening. Thus, CO induces significant alterations of cardiac electrical activity via cGMP-dependent mechanism and should be considered as a novel regulator of cardiac electrophysiology.
Collapse
|
47
|
Moustafa A, Habara Y. A novel role for carbon monoxide as a potent regulator of intracellular Ca2+and nitric oxide in rat pancreatic acinar cells. Am J Physiol Cell Physiol 2014; 307:C1039-49. [DOI: 10.1152/ajpcell.00252.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO) is known as an essential gaseous messenger that regulates a wide array of physiological and pathological processes, similar to nitric oxide (NO) and hydrogen sulfide. The aim of the present study was to elucidate the potential role of CO in Ca2+homeostasis and to explore the underlying mechanisms in pancreatic acinar cells. The exogenous application of a CO-releasing molecule dose-dependently increased intracellular Ca2+concentration ([Ca2+]i). A heme oxygenase (HO) inducer increased [Ca2+]iin a concentration-dependent manner, and the increase was diminished by an HO inhibitor. The CO-induced [Ca2+]iincrease persisted in the absence of extracellular Ca2+, indicating that Ca2+release is the initial source for the increase. The inhibition of G protein, phospholipase C (PLC), and inositol 1,4,5-trisphosphate (IP3) receptor diminished the CO-induced [Ca2+]iincrease. CO upregulated endothelial nitric oxide synthase (eNOS) expression and stimulated NO production, and NOS inhibitor, calmodulin inhibitor, or the absence of extracellular Ca2+eliminated the latter response. Blocking the phosphatidylinositol 3-kinase (PI3K)-Akt/protein kinase B (PKB) pathway abolished CO-induced NO production. Pretreatment with an NOS inhibitor, NO scavenger, or soluble guanylate cyclase inhibitor, did not affect the CO-induced [Ca2+]iincrease, indicating that NO, soluble guanylate cyclase, and cyclic guanosine 5′-monophosphate are not involved in the CO-induced [Ca2+]iincrease. CO inhibited the secretory responses to CCK-octapeptide or carbachol. We conclude that CO acts as a regulator not only for [Ca2+]ihomeostasis via a PLC-IP3-IP3receptor cascade but also for NO production via the calmodulin and PI3K-Akt/PKB pathway, and both CO and NO interact. Moreover, CO may provide potential therapy to ameliorate acute pancreatitis by inhibiting amylase secretion.
Collapse
Affiliation(s)
- Amira Moustafa
- Laboratory of Physiology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; and
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yoshiaki Habara
- Laboratory of Physiology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; and
| |
Collapse
|
48
|
Carbon monoxide is not always a poison gas for human organism: Physiological and pharmacological features of CO. Chem Biol Interact 2014; 222:37-43. [DOI: 10.1016/j.cbi.2014.08.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/02/2014] [Accepted: 08/18/2014] [Indexed: 01/21/2023]
|
49
|
Andreadou I, Iliodromitis EK, Rassaf T, Schulz R, Papapetropoulos A, Ferdinandy P. The role of gasotransmitters NO, H2S and CO in myocardial ischaemia/reperfusion injury and cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol 2014; 172:1587-606. [PMID: 24923364 DOI: 10.1111/bph.12811] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/02/2014] [Accepted: 06/06/2014] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease is one of the leading causes of morbidity and mortality worldwide. The development of cardioprotective therapeutic agents remains a partly unmet need and a challenge for both medicine and industry, with significant financial and social implications. Protection of the myocardium can be achieved by mechanical vascular occlusions such as preconditioning (PC), when brief episodes of ischaemia/reperfusion (I/R) are experienced prior to ischaemia; postconditioning (PostC), when the brief episodes are experienced at the immediate onset of reperfusion; and remote conditioning (RC), when the brief episodes are experienced in another vascular territory. The elucidation of the signalling pathways, which underlie the protective effects of PC, PostC and RC, would be expected to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent reperfusion injury. Gasotransmitters including NO, hydrogen sulphide (H2S) and carbon monoxide (CO) are a growing family of regulatory molecules that affect physiological and pathological functions. NO, H2S and CO share several common properties; they are beneficial at low concentrations but hazardous in higher amounts; they relax smooth muscle cells, inhibit apoptosis and exert anti-inflammatory effects. In the cardiovascular system, NO, H2S and CO induce vasorelaxation and promote cardioprotection. In this review article, we summarize current knowledge on the role of the gasotransmitters NO, H2S and CO in myocardial I/R injury and cardioprotection provided by conditioning strategies and highlight future perspectives in cardioprotection by NO, H2S, CO, as well as their donor molecules.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece
| | | | | | | | | | | |
Collapse
|
50
|
Finsterer J, Ohnsorge P. Influence of mitochondrion-toxic agents on the cardiovascular system. Regul Toxicol Pharmacol 2013; 67:434-45. [DOI: 10.1016/j.yrtph.2013.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 09/01/2013] [Accepted: 09/02/2013] [Indexed: 10/26/2022]
|