1
|
Mu Y, Yang X, Xie Y, Luo J, Wu S, Yang J, Zhao W, Chen J, Weng Y. Carbon monoxide-releasing Vehicle CO@TPyP-FeMOFs modulating macrophages phenotype in inflammatory wound healing. Nitric Oxide 2024; 149:49-59. [PMID: 38889652 DOI: 10.1016/j.niox.2024.06.005] [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: 03/29/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/20/2024]
Abstract
Healing of chronic wounds has been critically limited by prolonged inflammation. Carbon monoxide (CO) is a biologically active molecule with high potential based on its efficacy in modulating inflammation, promoting wound healing and tissue remodeling. Strategies to use CO as a gaseous drug to chronic wounds have emerged, but controlling the sustained release of CO at the wound site remains a major challenge. In this work, a porphyrin-Fe based metal organic frameworks, TPyP-FeMOFs was prepared. The synthesized TPyP-FeMOFs was high-temperature vacuum activated (AcTPyP-FeMOFs) and AcTPyP-FeMOFs had a relatively high Fe (II) content. CO sorption isotherms showed that AcTPyP-FeMOFs chemisorbed CO and thus CO release was sustained and prolonged. In vitro evaluation results showed that CO@TPyP-FeMOFs reduced the inflammatory level of lipopolysaccharide (LPS) activated macrophages, polarized macrophages to M2 anti-inflammatory phenotype, and promoted the proliferation of fibroblasts by altering the pathological microenvironment. In vivo study confirmed CO@TPyP-FeMOFs promoted healing in a LPS model of delayed cutaneous wound repair and reduced macrophages and neutrophils recruitment. Both in vitro and in vivo studies verified that CO@TPyP-FeMOFs acted on macrophages by modulating phenotype and inflammatory factor expression. Thus, CO release targeting macrophages and pathological microenvironment modulation presented a promising strategy for wound healing.
Collapse
Affiliation(s)
- Yixian Mu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xinlei Yang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yinhong Xie
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jie Luo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Sui Wu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - JinMing Yang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wei Zhao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Junying Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
2
|
Mansour AM, Khaled RM, Ferraro G, Shehab OR, Merlino A. Metal-based carbon monoxide releasing molecules with promising cytotoxic properties. Dalton Trans 2024; 53:9612-9656. [PMID: 38808485 DOI: 10.1039/d4dt00087k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Carbon monoxide, the "silent killer" gas, is increasingly recognised as an important signalling molecule in human physiology, which has beneficial biological properties. A particular way of achieving controlled CO administration is based on the use of biocompatible molecules that only release CO when triggered by internal or external factors. These approaches include the development of pharmacologically effective prodrugs known as CO releasing molecules (CORMs), which can supply biological systems with CO in well-regulated doses. An overview of transition metal-based CORMs with cytotoxic properties is here reported. The mechanisms at the basis of the biological activities of these molecules and their potential therapeutical applications with respect to their stability and CO releasing properties have been discussed. The activation of metal-based CORMs is determined by the type of metal and by the nature and features of the auxiliary ligands, which affect the metal core electronic density and therefore the prodrug resistance towards oxidation and CO release ability. A major role in regulating the cytotoxic properties of these CORMs is played by CO and/or CO-depleted species. However, several mysteries concerning the cytotoxicity of CORMs remain as intriguing questions for scientists.
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, United Arab Emirates University, Al-Ain, United Arab Emirates.
| | - Rabaa M Khaled
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt.
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| | - Ola R Shehab
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt.
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| |
Collapse
|
3
|
Bauer N, Yuan Z, Yang X, Wang B. Plight of CORMs: The unreliability of four commercially available CO-releasing molecules, CORM-2, CORM-3, CORM-A1, and CORM-401, in studying CO biology. Biochem Pharmacol 2023; 214:115642. [PMID: 37321416 PMCID: PMC10529722 DOI: 10.1016/j.bcp.2023.115642] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
Carbon monoxide (CO) is an endogenously produced gaseous signaling molecule with demonstrated pharmacological effects. In studying CO biology, three delivery forms have been used: CO gas, CO in solution, and CO donors of various types. Among the CO donors, four carbonyl complexes with either a transition metal ion or borane (BH3) (termed CO-releasing molecules or CORMs) have played the most prominent roles appearing in over 650 publications. These are CORM-2, CORM-3, CORM-A1, and CORM-401. Intriguingly, there have been unique biology findings that were only observed with these CORMs, but not CO gas; yet these properties were often attributed to CO, raising puzzling questions as to why CO source would make such a fundamental difference in terms of CO biology. Recent years have seen a large number of reports of chemical reactivity (e.g., catalase-like activity, reaction with thiol, and reduction of NAD(P)+) and demonstrated CO-independent biological activity for these four CORMs. Further, CORM-A1 releases CO in an idiosyncratic fashion; CO release from CORM-401 is strongly influenced or even dependent on reaction with an oxidant and/or a nucleophile; CORM-2 mostly releases CO2, not CO, after a water-gas shift reaction except in the presence of a strong nucleophile; and CORM-3 does not release CO except in the presence of a strong nucleophile. All these beg the question as to what constitutes an appropriate CO donor for studying CO biology. This review critically summarizes literature findings related to these aspects, with the aim of helping result interpretation when using these CORMs and development of essential criteria for an appropriate donor for studying CO biology.
Collapse
Affiliation(s)
- Nicola Bauer
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
4
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. The isothiocyanate sulforaphane prevents mitochondrial impairment and neuroinflammation in the human dopaminergic SH-SY5Y and in the mouse microglial BV2 cells: role for heme oxygenase-1. Metab Brain Dis 2023; 38:419-435. [PMID: 35469083 DOI: 10.1007/s11011-022-00990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 01/25/2023]
Abstract
Sulforaphane (SFN) promotes protective effects in different cell types. Nonetheless, it remains to be clarified by which mechanism SFN exerts benefits in mammalian cells. Mitochondria are a major source of adenosine triphosphate (ATP) and reactive species in nucleated cells. Mitochondrial impairment result in cellular redox biology disruption, bioenergetic status collapse, and inflammation. Evidence suggest that mitochondrial dysfunction plays a role in neurological disorders. Since a cure was not discovered yet to some of these diseases, investigating strategies to promote mitochondrial protection is pharmacologically relevant and may improve life quality of patients suffering from these maladies. Natural molecules, such as SFN, are potent inducers of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and, consequently, stimulate the expression of genes whose products, such as heme oxygenase-1 (HO-1), induce cytoprotective actions in mammalian tissues. In this work, we investigated whether SFN (5 µM) would be capable to prevent the dysfunctions caused by chlorpyrifos (CPF) on the human dopaminergic SH-SY5Y cells. Moreover, we examined the effects of a pretreatment with SFN at the same concentration on the mouse microglial BV2 cells stimulated by lipopolysaccharide (LPS) in an experimental model of neuroinflammation. SFN prevented the mitochondrial impairment and the neuroinflammation caused by the chemical stressors in both cell types. Inhibition of heme oxygenase-1 (HO-1) suppressed the mitochondrial protection and anti-inflammatory action afforded by SFN in this experimental model. Overall, SFN promoted cytoprotection by a mechanism dependent on the HO-1 enzyme in the SH-SY5Y and BV2 cells.
Collapse
Affiliation(s)
- Flávia Bittencourt Brasil
- Departamento de Ciências da Natureza, Campus Universitário de Rio das Ostras-Universidade Federal Fluminense (UFF), Rio de Janeiro, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Programa de Pós-Graduação Em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Matheus Dargesso Luckachaki
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Evandro Luiz Dall'Oglio
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil.
| |
Collapse
|
5
|
Bajia D, Bottani E, Derwich K. Effects of Noonan Syndrome-Germline Mutations on Mitochondria and Energy Metabolism. Cells 2022; 11:cells11193099. [PMID: 36231062 PMCID: PMC9563972 DOI: 10.3390/cells11193099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.
Collapse
Affiliation(s)
- Donald Bajia
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Piazzale L. A. Scuro 10, 37134 Verona, Italy
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| |
Collapse
|
6
|
Choi HI, Zeb A, Kim MS, Rana I, Khan N, Qureshi OS, Lim CW, Park JS, Gao Z, Maeng HJ, Kim JK. Controlled therapeutic delivery of CO from carbon monoxide-releasing molecules (CORMs). J Control Release 2022; 350:652-667. [PMID: 36063960 DOI: 10.1016/j.jconrel.2022.08.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 01/06/2023]
Abstract
Carbon monoxide (CO) has been regarded as a "silent killer" for its toxicity toward biological systems. However, a low concentration of endogenously produced CO has shown a number of therapeutic benefits such as anti-inflammatory, anti-proliferative, anti-apoptosis, and cytoprotective activities. Carbon monoxide-releasing molecules (CORMs) have been developed as alternatives to direct CO inhalation, which requires a specialized setting for strict dose control. CORMs are efficient CO donors, with central transition metals (such as ruthenium, iron, cobalt, and manganese) surrounded by CO as a ligand. CORMs can stably store and subsequently release their CO payload in the presence of certain triggers including solvent, light, temperature, and ligand substitution. However, CORMs require appropriate delivery strategies to improve short CO release half-life and target specificity. Herein, we highlighted the therapeutic potential of inhalation and CORMs-delivered CO. The applications of conjugate and nanocarrier systems for controlling CO release and improving therapeutic efficacy of CORMs are also described in detail. The review concludes with some of the hurdles that limit clinical translation of CORMs. Keeping in mind the tremendous potential and growing interest in CORMs, this review would be helpful for designing controlled CO release systems for clinical applications.
Collapse
Affiliation(s)
- Ho-Ik Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea
| | - Alam Zeb
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon, Republic of Korea; Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Min-Su Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea
| | - Isra Rana
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Namrah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Omer Salman Qureshi
- Department of Pharmacy, Faculty of Natural Sciences, Forman Christian College University, Lahore, Pakistan
| | - Chang-Wan Lim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea
| | - Jeong-Sook Park
- College of Pharmacy, Institute of Drug Research and Development, Chungnam National University, Daejeon, Republic of Korea
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon, Republic of Korea.
| | - Jin-Ki Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea.
| |
Collapse
|
7
|
Köktürk M, Yıldırım S, Eser G, Bulut M, Alwazeer D. Hydrogen-Rich Water Alleviates the Nickel-Induced Toxic Responses (Inflammatory Responses, Oxidative Stress, DNA Damage) and Ameliorates Cocoon Production in Earthworm. Biol Trace Elem Res 2022; 200:3442-3452. [PMID: 34482505 DOI: 10.1007/s12011-021-02908-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022]
Abstract
In recent years, studies investigating the protective effect of hydrogen-rich water (HRW) against different diseases and the toxicity of some substances have attracted increasing attention. Here, we assessed the effects of hydrogen-rich water on different nickel-induced toxic responses (reactive oxygen species (ROS), tumor necrosis factor-alpha (TNF-α), and 8-hydroxy-2'-deoxyguanosine (8-OHdG) of stress responses, histopathological changes) and cocoon production in earthworm model. Earthworms were randomly divided into two main groups: water (W) group including control (CW: ultrapure water), 10 (10W), 200 (200W), and 500 (500W), and hydrogen-rich ultrapure water (HRW) group including control (CHRW: hydrogen-rich ultrapure water), 10 (10HRW), 200 (200HRW), and 500 (500HRW) mg of nickel chloride kg-1 soil for 14 days. We found that cocoon production was less affected by the nickel exposure of earthworms in the 500HRW group compared to the 500W group. The ROS levels in 200HRW and 500HRW groups were less than that of 200W and 500W, respectively. The epithelial degeneration, epithelial necrosis, and necrosis in muscle fibers in tissues of earthworm were less damaged in 200HRW and 500HRW groups compared to 200W and 500W, respectively. HRW groups significantly reduced the expression of 8-OHdG induced by nickel exposure and inflammatory cytokine response including TNF-α. The study showed that hydrogen-rich water could alleviate the toxic effects of nickel-induced oxidative and inflammatory damages in earthworms. The HRW treatment known for its cheap and eco-friendly propertıes without any negative effects on the ecosystem can be used as a green method for alleviating the toxification effects of heavy metals in contaminated soil and increasing cocoon production of earthworms.
Collapse
Affiliation(s)
- Mine Köktürk
- Department of Organic Agriculture Management, College of Applied Sciences, Igdir University, Igdir, Turkey
| | - Serkan Yıldırım
- Department of Pathology, Faculty of Veterinary, Ataturk University, Erzurum, Turkey
| | - Gizem Eser
- Tuzluca Vocational School, Laboratory and Veterinary Health Programs, Igdir University, 76000 , Igdir, Turkey
| | - Menekşe Bulut
- Department of Food Engineering, Faculty of Engineering, Igdir University, 76000, Igdir, Turkey
- Research Center for Redox Applications in Foods (RCRAF), Igdir University, 76000 , Igdir, Turkey
- Innovative Food Technologies Development, Application, and Research Center, Igdir University, 76000, Igdir, Turkey
| | - Duried Alwazeer
- Research Center for Redox Applications in Foods (RCRAF), Igdir University, 76000 , Igdir, Turkey.
- Innovative Food Technologies Development, Application, and Research Center, Igdir University, 76000, Igdir, Turkey.
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Igdir University, 76000, Igdir, Turkey.
| |
Collapse
|
8
|
Cheng CY, Chen YH, Thuy Tien Vo T, Chui Hong Y, Wang CS, Canh Vo Q, Chou HC, Huang TW, Lee IT. CORM-2 prevents human gingival fibroblasts from lipoteichoic acid-induced VCAM-1 and ICAM-1 expression by inhibiting TLR2/MyD88/TRAF6/PI3K/Akt/ROS/NF-κB signaling pathway. Biochem Pharmacol 2022; 201:115099. [PMID: 35617999 DOI: 10.1016/j.bcp.2022.115099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/02/2022]
Abstract
Periodontal diseases are prevalent worldwide. Lipoteichoic acid (LTA), a major component of gram-positive bacteria, may play a key role in periodontally inflammatory diseases. Carbon monoxide (CO) is a critical messenger in many biological processes. It can elicit various biological properties, especially anti-inflammatory effects. As the straight administration of CO remains difficult, CO-releasing molecules (CO-RMs) are emerging as promising alternatives. To explore the pharmacological actions and signaling pathways of CO battling LTA-induced periodontal inflammation, this study investigated the cytoprotective effects of CORM-2 against the adhesion of THP-1 monocytes to human gingival fibroblasts (HGFs) and the underlying molecular mechanism. After exposing HGFs to LTA with or without CORM-2 pretreatment, monocyte adhesion was determined. VCAM-1 and ICAM-1 expression in HGFs was measured by real-time PCR. To identify the signaling pathways of CO involved in the cytoprotective effects of CORM-2, HGFs underwent pharmacological or genetical interventions before LTA incubation. The expression and/or activity of possible regulatory molecules were determined. The release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, were measured using ELISA. The results showed that LTA increased cytokine production and upregulated VCAM-1 and ICAM-1 expression in HGFs, promoting monocyte adhesion. These events were dependent on TLR2/MyD88/TRAF6- and PI3K/Akt/NADPH oxidase/ROS-regulated NF-κB activation. CORM-2 inhibited LTA-induced inflammatory cascades in HGFs, in which CO seemed to be the hitman. To conclude, CO released from CORM-2 can prevent the LTA-stimulated HGFs from increasing VCAM-1 and ICAM-1 expression and promoting monocyte adhesion by inhibiting TLR2/MyD88/TRAF6 association and PI3K/Akt/NADPH oxidase/ROS signaling, both converge on the canonical NF-κB activation.
Collapse
Affiliation(s)
- Ching-Yi Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, No. 261, Wenhua 1st Rd., Guishan Dist., Taoyuan City 333, Taiwan; Department of Pulmonary Infection and Immunology, Chang Gung Memorial Hospital at Linkou, No. 5, Fuxing St., Guishan Dist., Taoyuan City 333, Taiwan
| | - Yu-Hsu Chen
- Department of Orthopedic surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan; Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Thi Thuy Tien Vo
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying Chui Hong
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Quang Canh Vo
- Department of Dental Biomaterials Science, Dental Research Institute and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Han-Chin Chou
- Department of Chinese Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Ting-Wei Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Yan R, Ren J, Wen J, Cao Z, Wu D, Qin M, Xu D, Castillo R, Li F, Wang F, Gan Z, Liu C, Wei P, Lu Y. Enzyme Therapeutic for Ischemia and Reperfusion Injury in Organ Transplantation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105670. [PMID: 34617335 DOI: 10.1002/adma.202105670] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/18/2021] [Indexed: 06/13/2023]
Abstract
Ischemia-reperfusion injury (IRI) remains as a critical challenge for organ transplantation. Herein, an enzyme therapeutic based on superoxide dismutase and catalase for effective mitigation of IRI and pathogen-induced liver injury is reported, providing a therapeutic for organ transplantation and other diseases.
Collapse
Affiliation(s)
- Ran Yan
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jie Ren
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Di Wu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meng Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Duo Xu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Roxanne Castillo
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Feifei Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fang Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ping Wei
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
10
|
Yuan Z, Yang X, Wang B. Redox and catalase-like activities of four widely used carbon monoxide releasing molecules (CO-RMs). Chem Sci 2021; 12:13013-13020. [PMID: 34745532 PMCID: PMC8513939 DOI: 10.1039/d1sc03832j] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/05/2021] [Indexed: 12/30/2022] Open
Abstract
The pathophysiological roles of the endogenous signaling molecule, carbon monoxide (CO), have been extensively studied and validated in cell culture and animal models. Further, evidence supporting the therapeutic effects of CO in various human diseases has been mounting over the last two decades. Along this line, there has been intensive interest in developing various delivery forms including CO gas, CO in solution, metal–carbonyl complexes widely known as CO-releasing molecules (CO-RMs), and organic CO prodrugs. Among them, two ruthenium-based carbonyl complexes, CORM-2 and -3, occupy a very special place because they have been used in over 500 published studies. One of the mechanisms for CO's actions is known to be through attenuation of oxidative stress and regulation of production of reactive oxygen species (ROS). For this reason, it is important that CO delivery forms do not have intrinsic chemical redox properties. Herein, we describe our findings of catalase-like activities of CORM-2 and -3 in a CO-independent fashion, leading to the rapid degradation of hydrogen peroxide (H2O2) in PBS buffer (pH = 7.4) and in cell culture media. Further, we have found that CORM-2 and CORM-3 possess potent radical scavenging abilities. We have also studied two other widely used CO donors: CORM-401 and CORM-A1. Both showed chemical reactivity with ROS, but to a lesser degree than CORM-2 and -3. Because of the central role of ROS in some of the proposed mechanisms of actions for CO biology, the discovery of intrinsic chemical redox properties for these CO-RMs means that additional attention in designing proper controls is needed in future biological experiments using these CO-RMs for their CO-donating functions. Further, much more work is needed to understand the true implications of the chemical reactivity of these CO-RMs in cell-culture and animal-model studies of CO biology. Four CO-releasing molecules are found to degrade H2O2 and free radicals either catalytically (CORM-2 and -3) or through direct reactions (CORM-401 and -A1) in solution under near-physiological conditions.![]()
Collapse
Affiliation(s)
- Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| |
Collapse
|
11
|
CO-Releasing Molecule-2 Prevents Acute Kidney Injury through Suppression of ROS-Fyn-ER Stress Signaling in Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9947772. [PMID: 34326922 PMCID: PMC8277502 DOI: 10.1155/2021/9947772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) most commonly appears in critically ill patients in hospitals. AKI is characterized as a quick deterioration of kidney function and has recently been identified to be tightly interlinked with chronic kidney diseases. The emerging major mediators of AKI include oxidative stress and endoplasmic reticulum (ER) stress. Carbon monoxide (CO) attenuates oxidative stress and ER stress in various cells, while Fyn, a member of the Src kinase family, is activated by oxidative stress and contributes to ER stress in skeletal muscle. Considering these, the objective of the current research was to determine (i) the involvement of Fyn in ER stress-mediated AKI and (ii) the effect of CO-releasing molecule-2 (CORM2) on reactive oxygen species- (ROS-) Fyn-ER stress-mediated AKI. Pretreatment with CORM2 (30 mg/kg) efficiently inhibited LPS (30 mg/kg)-induced oxidative stress, inflammation, and cellular apoptosis during AKI in C57BL/6J mice. Also, CORM2 efficiently suppressed the activation of Fyn and ER stress in AKI mice. Consistently, pretreatment with CORM2 inhibited oxidative stress, Fyn activation, ER stress, inflammation, and apoptosis in LPS- or H2O2-stimulated proximal epithelial tubular cells. Fyn inhibition using siRNA or an inhibitor (PP2) significantly attenuated ER stress responses in the cells. These data suggest that CORM2 may become a potential treatment option against ROS-Fyn-ER stress-mediated AKI.
Collapse
|
12
|
García-Navas R, Liceras-Boillos P, Gómez C, Baltanás FC, Calzada N, Nuevo-Tapioles C, Cuezva JM, Santos E. Critical requirement of SOS1 RAS-GEF function for mitochondrial dynamics, metabolism, and redox homeostasis. Oncogene 2021; 40:4538-4551. [PMID: 34120142 PMCID: PMC8266680 DOI: 10.1038/s41388-021-01886-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/14/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
SOS1 ablation causes specific defective phenotypes in MEFs including increased levels of intracellular ROS. We showed that the mitochondria-targeted antioxidant MitoTEMPO restores normal endogenous ROS levels, suggesting predominant involvement of mitochondria in generation of this defective SOS1-dependent phenotype. The absence of SOS1 caused specific alterations of mitochondrial shape, mass, and dynamics accompanied by higher percentage of dysfunctional mitochondria and lower rates of electron transport in comparison to WT or SOS2-KO counterparts. SOS1-deficient MEFs also exhibited specific alterations of respiratory complexes and their assembly into mitochondrial supercomplexes and consistently reduced rates of respiration, glycolysis, and ATP production, together with distinctive patterns of substrate preference for oxidative energy metabolism and dependence on glucose for survival. RASless cells showed defective respiratory/metabolic phenotypes reminiscent of those of SOS1-deficient MEFs, suggesting that the mitochondrial defects of these cells are mechanistically linked to the absence of SOS1-GEF activity on cellular RAS targets. Our observations provide a direct mechanistic link between SOS1 and control of cellular oxidative stress and suggest that SOS1-mediated RAS activation is required for correct mitochondrial dynamics and function.
Collapse
Affiliation(s)
- Rósula García-Navas
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain
| | - Pilar Liceras-Boillos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain
| | - Carmela Gómez
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain
| | - Fernando C Baltanás
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain
| | - Nuria Calzada
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa3, (CSIC - Universidad Autónoma de Madrid), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa3, (CSIC - Universidad Autónoma de Madrid), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer - Enfermedades Raras (CIBERER), Madrid, Spain
| | - Eugenio Santos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC - Universidad de Salamanca), Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer - Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
13
|
Stucki D, Westhoff P, Brilhaus D, Weber APM, Brenneisen P, Stahl W. Carbon monoxide exposure activates ULK1 via AMPK phosphorylation in murine embryonic fibroblasts. INT J VITAM NUTR RES 2021; 93:122-131. [PMID: 34074127 DOI: 10.1024/0300-9831/a000714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Carbon monoxide (CO) is endogenously produced upon degradation of heme by heme oxygenases (HOs) and is suggested to act as a gaseous signaling molecule. The expression of HO-1 is triggered by the Nrf2-Keap1 signaling pathway which responds to exogenous stress signals and dietary constituents such as flavonoids and glucosinolates or reactive metabolic intermediates like 4-hydroxynonenal. Endogenous CO affects energy metabolism, regulates the utilization of glucose and addresses CYP450 enzymes. Using the CO releasing molecule-401 (CORM-401), we studied the effect of endogenous CO on ATP synthesis, AMP-signaling and activation of the AMPK pathway in cell culture. Upon exposure of cells to CORM-401, the mitochondrial ATP production rate was significantly decreased (P=0.007) to about 50%, while glycolytic ATP synthesis was unchanged (P=0.489). Total ATP levels were less affected as determined by mass spectrometry. Instead, levels of ADP and AMP were elevated following CORM-401 exposure by about two- (P=0.022) and four-fold (P=0.012) compared to control, respectively. Increased concentrations of AMP activate AMPK which was demonstrated by a 10 to 15-fold increased phosphorylation of Thr172 of the α-subunit of AMPK (P=0.025). A downstream target of AMPK is the kinase ULK1 which triggers autophagic and mitophagic processes. Activation of ULK1 after CO exposure was proven by a 3 to 5-fold elevated phosphorylation of ULK1 at Ser555 (P=0.004). The present data suggest that production of endogenous CO leads to increasing amounts of AMP which mediates AMPK-dependent downstream effects and likely triggers autophagic processes. Since dietary constituents and their metabolites induce the expression of the CO producing enzyme HO-1, CO signaling may also be involved in the cellular response to nutritional factors.
Collapse
Affiliation(s)
- David Stucki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Westhoff
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dominik Brilhaus
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas P M Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
14
|
Zhu J, Wang J, Wang G, Zhang J, Tao W, Liu C, Liu M, Zhang H, Xie R, Ye F, Liu Y, Fang W, Chen X, Li Y. Precise Identification of the Dimethyl Sulfoxide Triggered Tricarbonyldichlororuthenium(II) Dimer for Releasing CO. J Phys Chem Lett 2021; 12:4658-4665. [PMID: 33978423 DOI: 10.1021/acs.jpclett.1c00905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Low concentrations of carbon monoxide (CO) can play vital roles in pharmacological and physiological functions in the human body. The transition-metal carbonyl complexes of the tricarbonyldichlororuthenium(II) dimer [Ru2(CO)6Cl4 (CORM-2)] were proposed as CO-releasing molecules (CORMs) to improve the delivery efficiency of CO for therapeutic effects. The accurate identification of final products for CORMs in solution and the detailed mechanisms of the release of CO were the essential prerequisite for its effective physiological application, which have been deficient. In this study, utilizing the cutting-edge two-dimensional (2D) IR spectroscopy, with the intrinsic vibrational modes and the coupling information on dynamics of intramolecular vibrational energy redistribution (IVR), the final products of A, B, C, and E are accurately identified when CORM-2 is dissolved in dimethyl sulfoxide (DMSO). Furthermore, with the clues on intermolecular interaction and chemical exchange dynamics between different products, the transformations between different products are also directly characterized for the first time. These findings challenge the results from the classic 1D spectroscopic pattern, and they evidently demonstrated that the release of CO from CORM-2 in DMSO was slow and complicated with multiple reaction pathways. Combining with DFT simulations, the detailed mechanisms of release of CO for CORM-2 dissolved in DMSO are schematically proposed, which can significantly contribute to its drug optimization and pharmacological as well as physiological applications.
Collapse
Affiliation(s)
- Jiangrui Zhu
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juanjuan Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Guosheng Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jia Zhang
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Tao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Chang Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ming Liu
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Hao Zhang
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Ruipei Xie
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Songshan Lake Materials Laboratory, Chinese Academy of Sciences, Dongguan, Guangdong 523808, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Ying Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Weihai Fang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xuebo Chen
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yunliang Li
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Songshan Lake Materials Laboratory, Chinese Academy of Sciences, Dongguan, Guangdong 523808, China
| |
Collapse
|
15
|
Yuan Z, Yang X, Ye Y, Tripathi R, Wang B. Chemical Reactivities of Two Widely Used Ruthenium-Based CO-Releasing Molecules with a Range of Biologically Important Reagents and Molecules. Anal Chem 2021; 93:5317-5326. [PMID: 33745269 DOI: 10.1021/acs.analchem.1c00533] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ruthenium-based CO-releasing molecules (CO-RMs), CORM-2 and CORM-3, have been widely used as surrogates of CO for studying its biological effects in vitro and in vivo with much success. However, several previous solution-phase and in vitro studies have revealed the ability of such CO-RMs to chemically modify proteins and reduce aromatic nitro groups due to their intrinsic chemical reactivity under certain conditions. In our own work of studying the cytoprotective effects of CO donors, we were in need of assessing chemical factors that could impact the interpretation of results from CO donors including CORM-2,3 in various in vitro assays. For this, we examined the effects of CORM-2,3 toward representative reagents commonly used in various bioassays including resazurin, tetrazolium salts, nitrites, and azide-based H2S probes. We have also examined the effect of CORM-2,3 on glutathione disulfide (GSSG), which is a very important redox regulator. Our studies show the ability of these CO-RMs to induce a number of chemical and/or spectroscopic changes for several commonly used biological reagents under near-physiological conditions. These reactions/spectroscopic changes cannot be duplicated with CO-deleted CO-RMs (iCORMs), which are often used as negative controls. Furthermore, both CORM-2 and -3 are capable of consuming and reducing GSSG in solution. We hope that the results described will help in the future design of control experiments using Ru-based CO-RMs.
Collapse
Affiliation(s)
- Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Yuqian Ye
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Ravi Tripathi
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
16
|
Lu H, Merfeld-Clauss S, Jawed Y, March KL, Coleman ME, Bogatcheva NV. Distinct Factors Secreted by Adipose Stromal Cells Protect the Endothelium From Barrier Dysfunction and Apoptosis. Front Cell Dev Biol 2020; 8:584653. [PMID: 33102487 PMCID: PMC7554254 DOI: 10.3389/fcell.2020.584653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
We have shown previously that adipose stromal cell (ASC)-derived conditioned media (CM) limited lung injury, endothelial barrier dysfunction, and apoptosis. Here, we used endothelial hyperpermeability and apoptosis assays to investigate how concentration processes affect endothelium-directed bioactivity of ASC-CM and to gain information on the nature of bioactive factors. Comparison of ASC-CM concentrated with differential molecular weight (MW) cutoff filters showed that endothelial barrier protection depended on the species-specific factors in ASC-CM fractionated with MW > 50 kDa. Known barrier regulators-keratin growth factor (KGF), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF)-were detected in ASC-CM fraction of > 100 kDa. Pretreatment of endothelial monolayers with concentrations of KGF, VEGF, and HGF detected in ASC-CM showed that only KGF and HGF protect the endothelium from barrier dysfunction. Depletion of KGF and HGF from ASC-CM attenuated ASC-CM's ability to protect the endothelial barrier. In contrast to barrier-protective factors, apoptosis-protective factors fractionated with MW < 3 kDa and were not species-specific. Application of donors of apoptosis-mitigating gases showed that the CO donor carbon monoxide-releasing molecule 2 (CORM2) protected the endothelium from apoptosis, while the H2S donor NaSH did not. Knockdown of CO-generating heme oxygenase 1 in ASC attenuated ASC-CM's ability to protect the endothelium from apoptosis. We have shown that tumor necrosis factor alpha (TNFα)-induced apoptosis in endothelium is c-Jun N-terminal kinase (JNK)-dependent, and JNK activation is inhibited by ASC-CM pretreatment of endothelial cells. ASC-CM from heme oxygenase 1-depleted ASC displayed attenuated ability to suppress endothelial JNK activation, suggesting that CO-mediated protection of the endothelium from apoptosis is achieved by the downregulation of the JNK pathway. Altogether, our results demonstrate that the concentration of ASC-CM with low MW cutoff filters significantly reduces its anti-apoptotic activity while preserving its barrier-protective activity.
Collapse
Affiliation(s)
- Hongyan Lu
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Stephanie Merfeld-Clauss
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Yameena Jawed
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | - Keith L March
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States
| | | | - Natalia V Bogatcheva
- Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States.,Indiana Center for Vascular Biology and Medicine and Vascular and Cardiac Adult Stem Cell Therapy Center, Indianapolis, IN, United States.,Roudebush Veteran Affairs Medical Center, Indianapolis, IN, United States.,Division of Pulmonary, Sleep and Critical Care, Department of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
17
|
Li C, Chen L, Song M, Fang Z, Zhang L, Coffie JW, Zhang L, Ma L, Wang Q, Yang W, Fang L, Wang S, Gao X, Wang H. Ferulic acid protects cardiomyocytes from TNF-α/cycloheximide-induced apoptosis by regulating autophagy. Arch Pharm Res 2020; 43:863-874. [PMID: 32720163 DOI: 10.1007/s12272-020-01252-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/19/2020] [Indexed: 12/15/2022]
Abstract
Acute myocardial infarction (AMI) results in irreversible cardiac cell damage or death because of decreased blood flow to the heart. Apoptosis plays an important role in the process of tissue damage after myocardial infarction (MI), which has pathological and therapeutic implications. Ferulic acid (FA) is a phenolic acid endowed with strong antioxidative and cytoprotective activities. The present study aimed to investigate whether FA protects cardiomyocytes from apoptosis by regulating autophagy, which is a cellular self-digestion process, and one of the first lines of defense against oxidative stress. Apoptosis was induced by TNF-α (10 ng/mL) and cycloheximide (CHX, 5 μg/mL) in rat H9c2 cardiomyocytes. FA-inhibited TNF-α/CHX-induced apoptosis was determined by the quantification of TUNEL-positive cells, and the effect was associated with decreased ROS production and inhibited caspase3 activation. FA treatment enhanced autophagy and increased autophagy-associated protein expression, leading to an inhibition of mTOR signaling. When co-treated with 3-methyladenine (3-MA), an autophagy inhibitor, the anti-apoptotic effect of FA was attenuated. In an in vivo mouse MI model, FA treatment decreased the apoptotic cell number, reduced infarct size, and improved cardiac performance, as determined by histological and echocardiographic assessments. Taken collectively, these results suggest that FA could protect cardiomyocytes from apoptosis by enhancing autophagy.
Collapse
Affiliation(s)
- Chunxiao Li
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lu Chen
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Min Song
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhirui Fang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lusha Zhang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Joel Wake Coffie
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liyuan Zhang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lulu Ma
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qianyi Wang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenjie Yang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Leyu Fang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shaoxia Wang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd., West Area, Tuanbo New Town, Jinghai Dist., Tianjin, 301617, China
| | - Xiumei Gao
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hong Wang
- Tianji State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd., West Area, Tuanbo New Town, Jinghai Dist., Tianjin, 301617, China.
| |
Collapse
|
18
|
Stucki D, Steinhausen J, Westhoff P, Krahl H, Brilhaus D, Massenberg A, Weber APM, Reichert AS, Brenneisen P, Stahl W. Endogenous Carbon Monoxide Signaling Modulates Mitochondrial Function and Intracellular Glucose Utilization: Impact of the Heme Oxygenase Substrate Hemin. Antioxidants (Basel) 2020; 9:antiox9080652. [PMID: 32717801 PMCID: PMC7465082 DOI: 10.3390/antiox9080652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/22/2022] Open
Abstract
Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Heme oxygenase activity has been attributed to antioxidant defense via the redox cycling system of biliverdin and bilirubin. There is increasing evidence that CO is a gaseous signaling molecule and plays a role in the regulation of energy metabolism. Inhibitory effects of CO on the respiratory chain are well established, but the implication of such a process on the cellular stress response is not well understood. By means of extracellular flux analyses and isotopic tracing, we studied the effects of CO, either released from the CO donor CORM-401 or endogenously produced by heme oxygenases, on the respiratory chain and glucose metabolism. CORM-401 was thereby used as a tool to mimic endogenous CO production by heme oxygenases. In the long term (>60 min), CORM-401-derived CO exposure inhibited mitochondrial respiration, which was compensated by increased glycolysis accompanied by a loss of the ATP production rate and an increase in proton leakage. This effect pattern was likewise observed after endogenous CO production by heme oxygenases. However, in the present setting, these effects were only observed when sufficient substrate for heme oxygenases (hemin) was provided. Modulation of the HO-1 protein level was less important. The long-term influence of CO on glucose metabolism via glycolysis was preceded by a short-term response (<30 min) of the cells to CO. Stable isotope-labeling experiments and metabolic flux analysis revealed a short-term shift of glucose consumption from glycolysis to the pentose phosphate pathway (PPP) along with an increase in reactive oxygen species (ROS) generation. Overall, we suggest that signaling by endogenous CO stimulates the rapid formation of reduction equivalents (NADPH) via the PPP, and plays an additional role in antioxidant defense, e.g., via feed-forward stimulation of the bilirubin/biliverdin redox cycling system.
Collapse
Affiliation(s)
- David Stucki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Julia Steinhausen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Philipp Westhoff
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (P.W.); (D.B.)
| | - Heide Krahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Dominik Brilhaus
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (P.W.); (D.B.)
| | - Annika Massenberg
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Andreas P. M. Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany;
| | - Andreas S. Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
- Correspondence: ; Tel.: +49-211-811-2711
| |
Collapse
|
19
|
Juszczak M, Kluska M, Wysokiński D, Woźniak K. DNA damage and antioxidant properties of CORM-2 in normal and cancer cells. Sci Rep 2020; 10:12200. [PMID: 32699258 PMCID: PMC7376213 DOI: 10.1038/s41598-020-68948-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
In this study, we compared the effect of tricarbonyldichlororuthenium (II) dimer (CORM-2) and its CO-depleted molecule (iCORM-2) on human peripheral blood mononuclear cells (PBMCs) and human promyelocytic leukemia HL-60 cells. We determined cell viability, DNA damage and DNA repair kinetics. We also studied the effect of both compounds on DNA oxidative damage, free radical level and HO-1 gene expression. We showed that at low concentrations both CORM-2 and iCORM-2 stimulate PBMCs viability. After 24-h incubation, CORM-2 and iCORM-2, at the concentration of 100 µM, reduce the viability of both PBMCs and HL-60 cells. We also demonstrated that CORM-2 and iCORM-2, in the 0.01–100 µM concentration range, cause DNA damage such as strand breaks and alkaline labile sites. DNA damage was repaired efficiently only in HL-60 cells. CORM-2 significantly reduces oxidative stress induced by 1 mM H2O2 in normal and cancer cells. On the contrary, iCORM-2 in HL-60 cells increases the level of free radicals in the presence of 1 and 5 mM H2O2. We also revealed that both CORM-2 and iCORM-2 induce HO-1 gene expression. However, CORM-2 induces this gene to a greater extent than iCORM-2, especially in HL-60 cells at 100 µM. Finally, we showed that CORM-2 and iCORM-2 reduce H2O2-induced DNA oxidative damage. Furthermore, CORM-2 proved to be a compound with stronger antioxidant properties than iCORM-2. Our results suggest that both active CORM-2 and inactive iCORM-2 exert biological effects such as cyto- and genotoxicity, antioxidant properties and the ability to induce the HO-1 gene. The released CO as well as iCORM-2 can be responsible for these effects.
Collapse
Affiliation(s)
- Michał Juszczak
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Magdalena Kluska
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Daniel Wysokiński
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Katarzyna Woźniak
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|
20
|
Rotko D, Bednarczyk P, Koprowski P, Kunz WS, Szewczyk A, Kulawiak B. Heme is required for carbon monoxide activation of mitochondrial BK Ca channel. Eur J Pharmacol 2020; 881:173191. [PMID: 32422186 DOI: 10.1016/j.ejphar.2020.173191] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 11/24/2022]
Abstract
Carbon monoxide (CO) is an endogenously synthesized gaseous mediator and is involved in the regulation of numerous physiological processes. Mitochondria, in which hemoproteins are abundant, are among the targets for CO action. Large-conductance calcium-activated (mitoBKCa) channels in the inner mitochondrial membrane share multiple biophysical similarities with the BKCa channels of the plasma membrane and could be a potential target for CO. To test this hypothesis, the activity of the mitoBKCa channels in human astrocytoma U-87 MG cell mitochondria was assessed with the patch-clamp technique. The effects of CO-releasing molecules (CORMs), such as CORM-2, CORM-401, and CORM-A1, were compared to the application of a CO-saturated solution to the mitoBKCa channels in membrane patches. The applied CORMs showed pleiotropic effects including channel inhibition, while the CO-containing solution did not significantly modulate channel activity. Interestingly, CO applied to the mitoBKCa channels, which were inhibited by exogenously added heme, stimulated the channel. To summarize, our findings indicate a requirement of heme binding to the mitoBKCa channel for channel modulation by CO and suggest that CORMs might have complex unspecific effects on mitoBKCa channels.
Collapse
Affiliation(s)
- Daria Rotko
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pastuera 3, 02-093, Warsaw, Poland
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pastuera 3, 02-093, Warsaw, Poland
| | - Wolfram S Kunz
- Division of Neurochemistry, Department of Experimental Epileptology and Cognition Research University of Bonn, Sigmund-Freud Strasse 25, 53105, Bonn, Germany
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pastuera 3, 02-093, Warsaw, Poland
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pastuera 3, 02-093, Warsaw, Poland.
| |
Collapse
|
21
|
Stucki D, Krahl H, Walter M, Steinhausen J, Hommel K, Brenneisen P, Stahl W. Effects of frequently applied carbon monoxide releasing molecules (CORMs) in typical CO-sensitive model systems - A comparative in vitro study. Arch Biochem Biophys 2020; 687:108383. [PMID: 32335048 DOI: 10.1016/j.abb.2020.108383] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022]
Abstract
Intracellular carbon monoxide (CO) is a gaseous signaling molecule and is generated enzymatically by heme oxygenases upon degradation of heme to billiverdin. Target structures for intracellular produced CO are heme proteins including cytochrome c oxidase of the respiratory chain, cytochrome P450-dependent monooxygenases, or myoglobin. For studies on CO signaling, CO-releasing molecules (CORMs) of different structure are available. Here, three frequently used CORMs (CORM-2, CORM-3 and CORM-401) were studied for their properties to provide CO in biological test systems and address susceptible heme proteins. CO release was investigated in the myoglobin binding assay and found to be rapid (<5 min) with CORM-2- and CORM-3, whereas CORM-401 continuously provided CO (>50 min). Storage stability of CORM stock solutions was also assessed with the myoglobin assay. Only CORM-401 stock solutions were stable over a period of 7 days. Incubation of CORMs with recombinant cytochrome P450 led to an inhibition of enzyme activity. However, only CORM-3 and CORM-401 proved to be suitable in this test system because controls with the inactivated CORM-2 (iCORM-2) also led to a loss of enzyme activity. The impact of CORMs on the respiratory chain was investigated with high resolution respirometry and extracellular flux technology. In the first approach interferences of CORM-2 and CORM-3 with oxygen measurement occurred, since a rapid depletion of oxygen was detected in the medium even when no cells were present. However, CORM-401 did not interfere with oxygen measurement and the expected inhibition of cellular respiration was observed. CORM-2 was not suitable for use in oxygen measurements with the extracellular flux technology and CORM-3 application did not show any effect in this system. However, CO-dependent inhibition of cellular respiration was observed with CORM-401. Based on the present experiments it is concluded, that CORM-401 produced most reliable CO-specific results for the modulation of typical CO targets. For studies on CO-dependent biological effects on intracellular heme groups, CORM-2 and CORM-3 were less suitable. Depending on the experimental setting, data achieved with these compounds should be evaluated with caution.
Collapse
Affiliation(s)
- David Stucki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Heide Krahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Moritz Walter
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Julia Steinhausen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Katrin Hommel
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001, Düsseldorf, Germany.
| |
Collapse
|
22
|
Carbon Monoxide Being Hydrogen Sulfide and Nitric Oxide Molecular Sibling, as Endogenous and Exogenous Modulator of Oxidative Stress and Antioxidative Mechanisms in the Digestive System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5083876. [PMID: 32377300 PMCID: PMC7180415 DOI: 10.1155/2020/5083876] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/06/2020] [Accepted: 02/13/2020] [Indexed: 12/17/2022]
Abstract
Oxidative stress reflects an imbalance between oxidants and antioxidants in favor of the oxidants capable of evoking tissue damage. Like hydrogen sulfide (H2S) and nitric oxide (NO), carbon monoxide (CO) is an endogenous gaseous mediator recently implicated in the physiology of the gastrointestinal (GI) tract. CO is produced in mammalian tissues as a byproduct of heme degradation catalyzed by the heme oxygenase (HO) enzymes. Among the three enzymatic isoforms, heme oxygenase-1 (HO-1) is induced under conditions of oxidative stress or tissue injury and plays a beneficial role in the mechanism of protection against inflammation, ischemia/reperfusion (I/R), and many other injuries. According to recently published data, increased endogenous CO production by inducible HO-1, its delivery by novel pharmacological CO-releasing agents, or even the direct inhalation of CO has been considered a promising alternative in future experimental and clinical therapies against various GI disorders. However, the exact mechanisms underlying behind these CO-mediated beneficial actions are not fully explained and experimental as well as clinical studies on the mechanism of CO-induced protection are awaited. For instance, in a variety of experimental models related to gastric mucosal damage, HO-1/CO pathway and CO-releasing agents seem to prevent gastric damage mainly by reduction of lipid peroxidation and/or increased level of enzymatic antioxidants, such as superoxide dismutase (SOD) or glutathione peroxidase (GPx). Many studies have also revealed that HO-1/CO can serve as a potential defensive pathway against oxidative stress observed in the liver and pancreas. Moreover, increased CO levels after treatment with CO donors have been reported to protect the gut against formation of acute GI lesions mainly by the regulation of reactive oxygen species (ROS) production and the antioxidative activity. In this review, we focused on the role of H2S and NO molecular sibling, CO/HO pathway, and therapeutic potential of CO-releasing pharmacological tools in the regulation of oxidative stress-induced damage within the GI tract with a special emphasis on the esophagus, stomach, and intestines and also two solid and important metabolic abdominal organs, the liver and pancreas.
Collapse
|
23
|
Adach W, Błaszczyk M, Olas B. Carbon monoxide and its donors - Chemical and biological properties. Chem Biol Interact 2020; 318:108973. [PMID: 32035862 DOI: 10.1016/j.cbi.2020.108973] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/17/2020] [Accepted: 01/31/2020] [Indexed: 12/31/2022]
Abstract
Carbon monoxide (CO) is an inorganic chemical compound that can bind with hemoglobin with highly toxic effects. In living organisms, it is produced endogenously during the degradation of heme by oxygenase, which occurs in three isoforms: HO-1, HO-2 and HO-3. CO can play an important role in the regulation of many physiological functions. Carbon Oxide Releasing Molecules (CORMs) are a novel group of chemical compounds capable of controlled CO release directly in tissues or organs. This release depends on concentration, pH, solvent type and temperature. The biological role and the therapeutic potential of different CORMs is not always well demonstrated. However, this mini review summarizes the various function of these compounds.
Collapse
Affiliation(s)
- Weronika Adach
- University of Lodz, Department of General Biochemistry, Faculty of Biology and Environmental Protection, Pomorska 141/143, 90-236, Lodz, Poland
| | - Mateusz Błaszczyk
- University of Lodz, Department of General Biochemistry, Faculty of Biology and Environmental Protection, Pomorska 141/143, 90-236, Lodz, Poland
| | - Beata Olas
- University of Lodz, Department of General Biochemistry, Faculty of Biology and Environmental Protection, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|
24
|
Adach W, Olas B. A comparison of multifunctional donors of carbon monoxide: Their anticoagulant, antioxidant, anti-aggregatory and cytotoxicity activities in an in vitro model. Nitric Oxide 2020; 97:20-26. [PMID: 32006712 DOI: 10.1016/j.niox.2020.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/14/2019] [Accepted: 01/28/2020] [Indexed: 12/24/2022]
Abstract
The study examines the effect of two water-soluble carbon monoxide (CO) donors, CORM-3 and CORM-A1, on selected parameters of oxidative stress and hemostasis in human plasma and blood platelets in vitro. It also compares their activity with that of the lipid-soluble CORM-2. The oxidation of amino acid residues in plasma proteins was evaluated by measuring the amounts of thiol and carbonyl groups. Plasma lipid peroxidation was measured as thiobarbituric acid reactive substance (TBARS) concentration. In addition, three haemostatic parameters of plasma were studied, viz. activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT), and one haemostatic parameter of platelets (platelet aggregation). Treatment with CORM-3 and CORM-A1 (all concentrations from 0.1 to 100 μM) decreased thiol group oxidation induced by H2O2/Fe. Incubation with CORM-3 and CORM-A1 also influenced plasma coagulation activity, e.g. CORM-3 and CORM-A1 significantly prolonged TT at the two highest tested concentrations (50 and 100 μM). Only CORM-2 at the highest tested concentration (100 μM) and CORM-3 (50 and 100 μM) reduced platelet aggregation induced by ADP. None of the tested CORMs caused platelet damage. The treatment of various diseases associated with oxidative stress, including cardiovascular diseases, may be enhanced by the administration of CO donors CORM-2 and CORM-3, these being modulators of oxidative stress and hemostasis.
Collapse
Affiliation(s)
- Weronika Adach
- University of Lodz, Department of General Biochemistry, Faculty of Biology and Environmental Protection, Pomorska 141/143, 90-236, Lodz, Poland
| | - Beata Olas
- University of Lodz, Department of General Biochemistry, Faculty of Biology and Environmental Protection, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|
25
|
Walter M, Stahl W, Brenneisen P, Reichert AS, Stucki D. Carbon monoxide releasing molecule 401 (CORM-401) modulates phase I metabolism of xenobiotics. Toxicol In Vitro 2019; 59:215-220. [PMID: 31004742 DOI: 10.1016/j.tiv.2019.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/05/2019] [Accepted: 04/16/2019] [Indexed: 11/18/2022]
Abstract
Next to its well-studied toxicity, carbon monoxide (CO) is recognized as a signalling molecule in various cellular processes. Thus, CO-releasing molecules (CORMs) are of considerable interest for basic research and drug development. Aim of the present study was to investigate if CO, released from CORMs, inhibits cytochrome P450-dependent monooxygenase (CYP) activity and modulates xenobiotic metabolism. CORM-401 was used as a model CO delivering compound; inactive CORM-401 (iCORM-401), unable to release CO, served as control compound. CO release from CORM-401, but not from iCORM-401, was validated using the cell free myoglobin assay. CO-dependent inhibition of CYP activity was shown by 7-ethoxyresorufin-O-deethylation (EROD) with recombinant CYP and HepG2 cells. Upon CORM-401 exposure EROD activity of recombinant CYP decreased concentration dependently, while iCORM-401 had no effect. Treatment with CORM-401 decreased EROD activity in HepG2 cells at concentrations higher than 50 μM CORM-401, while iCORM-401 showed no effect. At the given concentrations cell viability was not affected. Amitriptyline was selected as a model xenobiotic and formation of its metabolite nortriptyline by recombinant CYP was determined by HPLC. CORM-401 treatment inhibited the formation of nortriptyline whereas iCORM-401 treatment did not. Overall, we demonstrate CO-mediated inhibitory effects on CYP activity when applying CORMs. Since CORMs are currently under drug development, the findings emphasize the importance to take into account that this class of compounds may interfere with xenobiotic metabolism.
Collapse
Affiliation(s)
- Moritz Walter
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001 Düsseldorf, Germany
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001 Düsseldorf, Germany.
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001 Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001 Düsseldorf, Germany
| | - David Stucki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Postfach 10 10 07, D-40001 Düsseldorf, Germany
| |
Collapse
|
26
|
Magierowska K, Bakalarz D, Wójcik D, Chmura A, Hubalewska-Mazgaj M, Licholai S, Korbut E, Kwiecien S, Sliwowski Z, Ginter G, Brzozowski T, Magierowski M. Time-dependent course of gastric ulcer healing and molecular markers profile modulated by increased gastric mucosal content of carbon monoxide released from its pharmacological donor. Biochem Pharmacol 2019; 163:71-83. [PMID: 30753813 DOI: 10.1016/j.bcp.2019.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/08/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND PURPOSE Besides hydrogen sulfide (H2S) and nitric oxide (NO), carbon monoxide (CO) contributes to the maintenance of gastric mucosal integrity. We investigated increased CO bioavailability effects on time-dependent dynamics of gastric ulcer healing mediated by particular growth factors, anti-inflammatory and molecular pathways. EXPERIMENTAL APPROACH Wistar rats with gastric ulcers induced by serosal acetic acid application (day 0) were treated i.g. throughout 3, 6 or 14 days with vehicle or CO-releasing tricarbonyldichlororuthenium (II) dimer (CORM-2, 2.5 mg/kg). Gross and microscopic alterations in gastric ulcer size and gastric blood flow (GBF) at ulcer margin were determined by planimetry, histology and laser flowmetry, respectively. Gastric mRNA/protein expressions of platelet derived growth factors (PDGFA-D), insulin-like growth factor (IGF-1), epidermal growth factor (EGF), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGFA) and their receptors, heme oxygenases (HMOX), nuclear factor (erythroid-derived 2)-like 2 (Nrf-2), cyclooxygenase (COX-2), hypoxia inducible factor (HIF)-1α, anti-inflammatory annexin-1 and transforming growth factor (TGF-β1) were assessed by real-time PCR or Western blot. TGF-β1-3 and IL-10 plasma concentration were measured using Luminex platform. Prostaglandin E2 content at ulcer margin was assessed by ELISA. KEY RESULTS CORM-2 decreased ulcer area and increased GBF after 6 and 14 days of treatment comparing to vehicle. CO donor upregulated HGF, HGFr, VEGFR1, VEGFR2, TGF-β1, annexin-1 and maintained increased IGF-1, PDGFC and EGF expression at various time-intervals of ulcer healing. TGF-β3 and IL-10 plasma concentration were significantly increased after COMR-2 vs. vehicle. CONCLUSIONS CO time-dependently accelerates gastric ulcer healing and raises GBF at ulcer margin by mechanism involving subsequent upregulation of anti-inflammatory, growth promoting and angiogenic factors response, not observed physiologically.
Collapse
Affiliation(s)
- Katarzyna Magierowska
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, 9 Westerplatte Street, 31-033 Cracow, Poland
| | - Dagmara Wójcik
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Anna Chmura
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Magdalena Hubalewska-Mazgaj
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Sabina Licholai
- Department of Molecular Biology and Clinical Genetics, Jagiellonian University Medical College, 8 Skawinska Street, 31-066 Cracow, Poland
| | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Slawomir Kwiecien
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Zbigniew Sliwowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Grzegorz Ginter
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland.
| |
Collapse
|
27
|
Soboleva T, Esquer HJ, Anderson SN, Berreau LM, Benninghoff AD. Mitochondrial-Localized Versus Cytosolic Intracellular CO-Releasing Organic PhotoCORMs: Evaluation of CO Effects Using Bioenergetics. ACS Chem Biol 2018; 13:2220-2228. [PMID: 29932318 PMCID: PMC6117112 DOI: 10.1021/acschembio.8b00387] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
While interactions between carbon monoxide (CO) and mitochondria have been previously studied, the methods used to deliver CO (gas or CO-releasing metal carbonyl compounds) lack subcellular targeting and/or controlled delivery. Thus, the effective concentration needed to produce changes in mitochondrial bioenergetics is yet to be fully defined. To evaluate the influence of mitochondrial-targeted versus intracellularly released CO on mitochondrial oxygen consumption rates, we developed and characterized flavonol-based CO donor compounds that differ at their site of release. These molecules are metal-free, visible light triggered CO donors (photoCORMs) that quantitatively release CO and are trackable in cells via confocal microscopy. Our studies indicate that at a concentration of 10 μM, the mitochondrial-localized and cytosolic CO-releasing compounds are similarly effective in terms of decreasing ATP production, maximal respiration, and the reserve capacity of A549 cells. This concentration is the lowest to impart changes in mitochondrial bioenergetics for any CO-releasing molecule (CORM) reported to date. The results reported herein demonstrate the feasibility of using a structurally tunable organic photoCORM framework for comparative intracellular studies of the biological effects of carbon monoxide.
Collapse
Affiliation(s)
- Tatiana Soboleva
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Hector J. Esquer
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah 84322-4815, United States
| | - Stacey N. Anderson
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Lisa M. Berreau
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Abby D. Benninghoff
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah 84322-4815, United States
| |
Collapse
|
28
|
Emerging role of carbon monoxide in regulation of cellular pathways and in the maintenance of gastric mucosal integrity. Pharmacol Res 2018; 129:56-64. [DOI: 10.1016/j.phrs.2018.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/12/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022]
|
29
|
Adach W, Olas B. The role of CORM-2 as a modulator of oxidative stress and hemostatic parameters of human plasma in vitro. PLoS One 2017; 12:e0184787. [PMID: 28950024 PMCID: PMC5614530 DOI: 10.1371/journal.pone.0184787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The main aim of the experiment is to examine the effect of CORM-2, a donor of carbon monoxide (CO), on oxidative stress in human plasma in vitro. In addition, it examines the effects of CORM-2 on the hemostatic parameters of plasma: the activated partial thromboplastin time (APTT), thrombin time (TT) and prothrombin time (PT). METHODS Human plasma was incubated for 5-60 min with different concentrations of CORM-2: 0.1-100 μM. Following this, various hemostatic factors and biomarkers of oxidative stress were studied. Lipid peroxidation was measured as thiobarbituric acid reactive substance (TBARS) concentration, and the oxidation of amino acid residues in proteins was measured by determining the amounts of carbonyl and thiol groups. RESULTS Two oxidative stress inducers: hydrogen peroxide (H2O2) and the donor of hydroxyl radical (H2O2/Fe) were used. Decrease in protein carbonylation, thiol group oxidation and lipid peroxidation were detected at tested concentrations of CORM-2. CONCLUSION Our results indicate that CORM-2 may have antioxidant properties in human plasma treated with H2O2 or H2O2/Fe. In addition, our results indicate the anti-coagulant activities of CORM-2 in vitro.
Collapse
Affiliation(s)
- Weronika Adach
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- * E-mail:
| |
Collapse
|