1
|
Mansoori R, Ashrafpour M, Asghari MH, Golchoobian R, Hosseini SM, Reiter RJ, Karim B, Moghadamnia AA, Kazemi S. Protective effects of melatonin against 5-fluorouracil-induced cardiotoxicity in rats: A comprehensive evaluation of oxidative, inflammatory, and apoptotic pathways. Toxicol Appl Pharmacol 2025; 499:117343. [PMID: 40239743 DOI: 10.1016/j.taap.2025.117343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Cardiotoxicity is a serious adverse effect of 5-fluorouracil (5-FU) a common chemotherapeutic agent. This study aimed to evaluate the protective effects of melatonin (MLT) against 5-fluorouracil (5-FU)-induced cardiotoxicity in rats, focusing on oxidative stress, inflammatory pathways, gene expression, electrocardiographic and histopathological changes. MATERIALS AND METHODS Twenty-five male Wistar rats were divided into five groups. The animals received either MLT at doses of 2.5, 5, or 10 mg/kg/day, 5-FU at 50 mg/kg (i.p.), or a combination of both treatments. Cardiotoxicity was assessed through electrocardiography, cardiac enzymes, oxidative stress markers, and histopathology. RESULTS 5-FU treatment significantly increased oxidative stress markers and inflammatory mediators while causing histopathological damage in heart tissues. Co-administration of MLT with 5-FU significantly mitigated these effects by reducing oxidative damage, as evidenced by lower levels of malondialdehyde (MDA), nitric oxide (NO), and myeloperoxidase (MPO). Additionally, MLT enhanced antioxidant activity, as reflected by increased levels of superoxide dismutase (SOD) and glutathione peroxidase (GPx) in heart tissues. Gene expression analysis further confirmed that MLT treatment reduced the elevated levels of COX-2 and VEGF, which are critical players in the inflammatory process. Histopathological examination demonstrated that MLT preserved the structural integrity of myocardial tissues, reducing 5-FU-induced damage score in a dose-dependent manner. Furthermore, MLT co-administration significantly attenuated the rise in cardiac biomarkers, including LDH, AST, and CK-MB, associated with 5-FU-induced cardiotoxicity. CONCLUSION These findings highlight that MLT, through its antioxidant and anti-inflammatory properties, exerts a protective effect against 5-FU-induced toxicity, suggesting its therapeutic potential for improving cardiovascular health during chemotherapy.
Collapse
Affiliation(s)
- Razieh Mansoori
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Manoochehr Ashrafpour
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Hossien Asghari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ravieh Golchoobian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
2
|
Zhi W, Li Y, Wang L, Hu X. Advancing Neuroscience and Therapy: Insights into Genetic and Non-Genetic Neuromodulation Approaches. Cells 2025; 14:122. [PMID: 39851550 PMCID: PMC11763439 DOI: 10.3390/cells14020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Neuromodulation stands as a cutting-edge approach in the fields of neuroscience and therapeutic intervention typically involving the regulation of neural activity through physical and chemical stimuli. The purpose of this review is to provide an overview and evaluation of different neuromodulation techniques, anticipating a clearer understanding of the future developmental trajectories and the challenges faced within the domain of neuromodulation that can be achieved. This review categorizes neuromodulation techniques into genetic neuromodulation methods (including optogenetics, chemogenetics, sonogenetics, and magnetogenetics) and non-genetic neuromodulation methods (including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, transcranial ultrasound stimulation, photobiomodulation therapy, infrared neuromodulation, electromagnetic stimulation, sensory stimulation therapy, and multi-physical-factor stimulation techniques). By systematically evaluating the principles, mechanisms, advantages, limitations, and efficacy in modulating neuronal activity and the potential applications in interventions of neurological disorders of these neuromodulation techniques, a comprehensive picture is gradually emerging regarding the advantages and challenges of neuromodulation techniques, their developmental trajectory, and their potential clinical applications. This review highlights significant advancements in applying these techniques to treat neurological and psychiatric disorders. Genetic methods, such as sonogenetics and magnetogenetics, have demonstrated high specificity and temporal precision in targeting neuronal populations, while non-genetic methods, such as transcranial magnetic stimulation and photobiomodulation therapy, offer noninvasive and versatile clinical intervention options. The transformative potential of these neuromodulation techniques in neuroscience research and clinical practice is underscored, emphasizing the need for integration and innovation in technologies, the optimization of delivery methods, the improvement of mediums, and the evaluation of toxicity to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Ying Li
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| |
Collapse
|
3
|
Shabanian K, Shabanian T, Karsai G, Pontiggia L, Paneni F, Ruschitzka F, Beer JH, Saeedi Saravi SS. AQP1 differentially orchestrates endothelial cell senescence. Redox Biol 2024; 76:103317. [PMID: 39180980 PMCID: PMC11388013 DOI: 10.1016/j.redox.2024.103317] [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: 03/18/2024] [Revised: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Accumulation of senescent endothelial cells (ECs) with age is a pivotal driver of cardiovascular diseases in aging. However, little is known about the mechanisms and signaling pathways that regulate EC senescence. In this report, we delineate a previously unrecognized role of aquaporin 1 (AQP1) in orchestrating extracellular hydrogen peroxide (H2O2)-induced cellular senescence in aortic ECs. Our findings underscore AQP1's differential impact on senescence hallmarks, including cell-cycle arrest, senescence-associated secretory phenotype (SASP), and DNA damage responses, intricately regulating angiogenesis. In proliferating ECs, AQP1 is crucial for maintaining angiogenic capacity, whereas disruption of AQP1 induces morphological and mitochondrial alterations, culminating in senescence and impaired angiogenesis. Conversely, Aqp1 knockdown or selective blockade of AQP1 in senescent ECs rescues the excess H2O2-induced cellular senescence phenotype and metabolic dysfunction, thereby ameliorating intrinsic angiogenic incompetence. Mechanistically, AQP1 facilitates H2O2 transmembrane transport, exacerbating oxidant-sensitive kinases CaMKII-AMPK. This process suppresses HDAC4 translocation, consequently de-repressing Mef2A-eNOS signaling in proliferating ECs. However, in senescent ECs, AQP1 overexpression is linked to preserved HDAC4-Mef2A complex and downregulation of eNOS signaling. Together, our studies identify AQP1 as a novel epigenetic regulator of HDAC4-Mef2A-dependent EC senescence and angiogenic potential, highlighting its potential as a therapeutic target for antagonizing age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, 8952, Schlieren, Switzerland
| | - Luca Pontiggia
- Tissue Biology Research Unit, University Children's Hospital Zurich, 8952, Schlieren, Switzerland; Children's Research Center, University Children's Hospital Zurich, 8032, Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404, Baden, Switzerland.
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Altun HY, Secilmis M, Yang F, Akgul Caglar T, Vatandaslar E, Toy MF, Vilain S, Mann GE, Öztürk G, Eroglu E. Visualizing hydrogen peroxide and nitric oxide dynamics in endothelial cells using multispectral imaging under controlled oxygen conditions. Free Radic Biol Med 2024; 221:89-97. [PMID: 38735541 DOI: 10.1016/j.freeradbiomed.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/04/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The complex interplay between hydrogen peroxide (H2O2) and nitric oxide (NO) in endothelial cells presents challenges due to technical limitations in simultaneous measurement, hindering the elucidation of their direct relationship. Previous studies have yielded conflicting findings regarding the impact of H2O2 on NO production. To address this problem, we employed genetically encoded biosensors, HyPer7 for H2O2 and geNOps for NO, allowing simultaneous imaging in single endothelial cells. Optimization strategies were implemented to enhance biosensor performance, including camera binning, temperature regulation, and environmental adjustments to mimic physiological normoxia. Our results demonstrate that under ambient oxygen conditions, H2O2 exhibited no significant influence on NO production. Subsequent exploration under physiological normoxia (5 kPa O2) revealed distinct oxidative stress levels characterized by reduced basal HyPer7 signals, enhanced H2O2 scavenging kinetics, and altered responses to pharmacological treatment. Investigation of the relationship between H2O2 and NO under varying oxygen conditions revealed a lack of NO response to H2O2 under hyperoxia (18 kPa O2) but a modest NO response under physiological normoxia (5 kPa O2). Importantly, the NO response was attenuated by l-NAME, suggesting activation of eNOS by endogenous H2O2 generation upon auranofin treatment. Our study highlights the intricate interplay between H2O2 and NO within the endothelial EA.hy926 cell line, emphasizing the necessity for additional research within physiological contexts due to differential response observed under physiological normoxia (5 kPa O2). This further investigation is essential for a comprehensive understanding of the H2O2 and NO signaling considering the physiological effects of ambient O2 levels involved.
Collapse
Affiliation(s)
- Hamza Yusuf Altun
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Melike Secilmis
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Fan Yang
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Tuba Akgul Caglar
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Emre Vatandaslar
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Muhammed Fatih Toy
- School of Engineering and Natural Science, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Sven Vilain
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Gürkan Öztürk
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Physiology Department, School of Medicine, Bolu Abant Izzet Baysal University (BAIBU), 14200, Bolu, Türkiye.
| | - Emrah Eroglu
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey.
| |
Collapse
|
5
|
Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GES. Possible role of LCZ696 in atherosclerosis: new inroads and perspective. Mol Cell Biochem 2024; 479:1895-1908. [PMID: 37526794 DOI: 10.1007/s11010-023-04816-x] [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: 04/14/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023]
Abstract
LCZ696 blocks both angiotensin receptor type 1 (ATR1) and neprilysin (NEP), which are intricate in the degradation of natriuretic peptides (NPs) and other endogenous peptides. It has been shown NEP inhibitors and LCZ696 could be effectively in the management of atherosclerosis (AS). However, the underlying mechanism of LCZ696 in AS is needed to be clarified entirely. Hence, this review is directed to reconnoiter the mechanistic role of LCZ696 in AS. The anti-inflammatory role of LCZ696 is related to the inhibition of transforming growth factor beta (TGF-β)-activated kinase 1 (TAK) and nod-like receptor pyrin 3 receptor (NLRP3) inflammasome. Moreover, LCZ696, via inhibition of pro-inflammatory cytokines, oxidative stress, apoptosis and endothelial dysfunction can attenuate the development and progression of AS. In conclusion, LCZ696 could be effective in the management of AS through modulation of inflammatory and oxidative signaling. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, Baghdad, Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AL Beheira, Egypt.
| |
Collapse
|
6
|
Guo R, Spyropoulos F, Michel T. FRBM Mini REVIEW: Chemogenetic approaches to probe redox dysregulation in heart failure. Free Radic Biol Med 2024; 217:173-178. [PMID: 38565399 PMCID: PMC11221410 DOI: 10.1016/j.freeradbiomed.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
Chemogenetics refers to experimental methods that use novel recombinant proteins that can be dynamically and uniquely regulated by specific biochemicals. Chemogenetic approaches allow the precise manipulation of cellular signaling to delineate the molecular pathways involved in both physiological and pathological disease states. Approaches utilizing yeast d-amino acid oxidase (DAAO) enable manipulation of intracellular redox metabolism through generation of hydrogen peroxide in the presence of d-amino acids and have led to the development of new and informative animal models to characterize the impact of oxidative stress in heart failure and neurodegeneration. These chemogenetic models, in which DAAO expression is regulated by different tissue-specific promoters, have led to a range of cardiac phenotypes. This review discusses chemogenetic approaches to manipulate oxidative stress in models of heart failure. These approaches provide new insights into the relationships between redox metabolism and normal and pathologic states in the heart, as well as in other diseases characterized by oxidative stress.
Collapse
Affiliation(s)
- Ruby Guo
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 02115, USA
| | - Fotios Spyropoulos
- Newborn Medicine Division, Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, USA
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 02115, USA.
| |
Collapse
|
7
|
Spyropoulos F, Michel T. D-Amino acid oxidase-derived chemogenetic oxidative stress: Unraveling the multi-omic responses to in vivo redox stress. Curr Opin Chem Biol 2024; 79:102438. [PMID: 38417321 PMCID: PMC10957096 DOI: 10.1016/j.cbpa.2024.102438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2024]
Abstract
Chemogenetic approaches have been developed to define the mechanisms whereby the intracellular oxidant hydrogen peroxide (H2O2) modulates both physiological and pathological responses. Recombinant yeast D-amino acid oxidase (DAAO) can be exploited to modulate H₂O₂ in target cells and tissues. In vitro studies using cultured cells expressing recombinant DAAO have provided critical new information on the intracellular transport and metabolism of H2O2 with great temporal and spatial resolution. In contrast, in vivo studies using chemogenetic/transgenic animal models have explored the pathological effects of chronically elevated H2O2 in tissues. Coupled with transcriptomic, proteomic, and metabolomic methods, in vivo chemogenetic approaches are providing new insights into the adaptations to oxidative stress. This review of chemogenetic applications focuses on new models of heart failure and neurodegeneration that leverage in vivo chemogenetic modulation of oxidative stress in target tissues to identify new therapeutic targets.
Collapse
Affiliation(s)
- Fotios Spyropoulos
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, USA.
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 02115, USA.
| |
Collapse
|
8
|
Spyropoulos F, Das AA, Waldeck-Weiermair M, Yadav S, Pandey AK, Guo R, Covington TA, Thulabandu V, Kosmas K, Steinhorn B, Perrella MA, Liu X, Christou H, Michel T. Adult and neonatal models of chemogenetic heart failure caused by oxidative stress. J Clin Invest 2024; 134:e178251. [PMID: 38483521 PMCID: PMC11060721 DOI: 10.1172/jci178251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Affiliation(s)
| | | | | | - Shambhu Yadav
- Division of Cardiovascular Medicine, Department of Medicine; and
| | - Arvind K. Pandey
- Division of Cardiovascular Medicine, Department of Medicine; and
| | - Ruby Guo
- Division of Cardiovascular Medicine, Department of Medicine; and
| | | | | | - Kosmas Kosmas
- Division of Newborn Medicine, Department of Pediatrics
| | | | - Mark A. Perrella
- Division of Newborn Medicine, Department of Pediatrics
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoli Liu
- Division of Newborn Medicine, Department of Pediatrics
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine; and
| |
Collapse
|
9
|
Li G, Liu X, Long A, Feng J, Sun S, Yang Z, Jiang R, Jiang X. An inducible mouse model of heart failure targeted to cardiac Drd5 deficiency detonating mitochondrial oxidative stress. Int J Cardiol 2024; 396:131560. [PMID: 38164957 DOI: 10.1016/j.ijcard.2023.131560] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 01/03/2024]
Affiliation(s)
- Guoqing Li
- The Affiliated Bozhou Hospital of Anhui Medical University (Bozhou People's Hospital), 616 Duzhong Road, Bozhou 236800, PR China
| | - Xing Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China
| | - Anxiong Long
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China
| | - Jianghao Feng
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China
| | - Shiyun Sun
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China
| | - Rongyan Jiang
- The Affiliated Bozhou Hospital of Anhui Medical University (Bozhou People's Hospital), 616 Duzhong Road, Bozhou 236800, PR China.
| | - Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, PR China.
| |
Collapse
|
10
|
Allemann MS, Lee P, Beer JH, Saeedi Saravi SS. Targeting the redox system for cardiovascular regeneration in aging. Aging Cell 2023; 22:e14020. [PMID: 37957823 PMCID: PMC10726899 DOI: 10.1111/acel.14020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.
Collapse
Affiliation(s)
- Meret Sarah Allemann
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Pratintip Lee
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of CardiologyUniversity Hospital Zurich, University of ZurichSchlierenSwitzerland
| |
Collapse
|
11
|
den Toom WTF, van Soest DMK, Polderman PE, van Triest MH, Bruurs LJM, De Henau S, Burgering BMT, Dansen TB. Oxygen-consumption based quantification of chemogenetic H 2O 2 production in live human cells. Free Radic Biol Med 2023; 206:134-142. [PMID: 37392950 DOI: 10.1016/j.freeradbiomed.2023.06.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Reactive Oxygen Species (ROS) in the form of H2O2 can act both as physiological signaling molecules as well as damaging agents, depending on their concentration and localization. The downstream biological effects of H2O2 were often studied making use of exogenously added H2O2, generally as a bolus and at supraphysiological levels. But this does not mimic the continuous, low levels of intracellular H2O2 production by for instance mitochondrial respiration. The enzyme d-Amino Acid Oxidase (DAAO) catalyzes H2O2 formation using d-amino acids, which are absent from culture media, as a substrate. Ectopic expression of DAAO has recently been used in several studies to produce inducible and titratable intracellular H2O2. However, a method to directly quantify the amount of H2O2 produced by DAAO has been lacking, making it difficult to assess whether observed phenotypes are the result of physiological or artificially high levels of H2O2. Here we describe a simple assay to directly quantify DAAO activity by measuring the oxygen consumed during H2O2 production. The oxygen consumption rate (OCR) of DAAO can directly be compared to the basal mitochondrial respiration in the same assay, to estimate whether the ensuing level of H2O2 production is within the range of physiological mitochondrial ROS production. In the tested monoclonal RPE1-hTERT cells, addition of 5 mM d-Ala to the culture media amounts to a DAAO-dependent OCR that surpasses ∼5% of the OCR that stems from basal mitochondrial respiration and hence produces supra-physiological levels of H2O2. We show that the assay can also be used to select clones that express differentially localized DAAO with the same absolute level of H2O2 production to be able to discriminate the effects of H2O2 production at different subcellular locations from differences in total oxidative burden. This method therefore greatly improves the interpretation and applicability of DAAO-based models, thereby moving the redox biology field forward.
Collapse
Affiliation(s)
- Wytze T F den Toom
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Daan M K van Soest
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Paulien E Polderman
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Miranda H van Triest
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Lucas J M Bruurs
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Sasha De Henau
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands
| | - Boudewijn M T Burgering
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands; Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, the Netherlands
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands.
| |
Collapse
|
12
|
Saravi SSS, Bonetti NR, Vukolic A, Vdovenko D, Lee P, Liberale L, Basso C, Rizzo S, Akhmedov A, Lüscher TF, Camici GG, Beer JH. Long-term dietary n3 fatty acid prevents aging-related cardiac diastolic and vascular dysfunction. Vascul Pharmacol 2023; 150:107175. [PMID: 37105373 DOI: 10.1016/j.vph.2023.107175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
AIMS The prevalence of left ventricular (LV) diastolic and vascular dysfunction increases with age, eventually leading to heart failure with preserved ejection fraction (HFpEF). A preventive strategy is an unmet medical need. We and others reported previously on the beneficial effects of omega-3 fatty acid alpha linolenic acid (ALA) on cardiovascular disorders in animal models and translational studies. We now investigate whether long-term dietary ALA could prevent LV diastolic dysfunction and vascular aging in a murine model. METHODS AND RESULTS Wild-type C57BL/6 J mice were fed a chow or ALA diet for 12 months, starting at 6 months of age. Here, we show that aged (~18 months) mice recapitulate major hallmarks of HFpEF, including LV diastolic dysfunction with preserved ejection fraction, impaired vascular function, cardiac fibrosis, arterial stiffening and inflammation, as well as elevated B-type natriuretic peptide (BNP). Long-term ALA supplementation upregulated the mitochondrial tricarboxylic acid enzyme Idh2 and the antioxidant enzymes SOD1 and Gpx1. It also has been associated with reduced inflammation and ECM remodeling, accompanied by a significant downregulation of fibrosis biomarkers MMP-2 and TGF-β in both cardiac and vascular tissues obtained from aged mice. Our data exhibited the preventive effects of dietary ALA against LV diastolic dysfunction, impaired vasorelaxation, cardiac fibrosis, inflammation and arterial stiffening in aged mice. CONCLUSIONS We provide evidence and a simplified mechanistic insight on how long-term ALA supplementation is a successful strategy to prevent the development of age-related diastolic and vascular dysfunction.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
| | - Nicole R Bonetti
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland
| | - Ana Vukolic
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Pratintip Lee
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Stefania Rizzo
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Royal Brompton and Harefield Hospitals, Imperial and Kings College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland.
| |
Collapse
|
13
|
Ponzoni M, Coles JG, Maynes JT. Rodent Models of Dilated Cardiomyopathy and Heart Failure for Translational Investigations and Therapeutic Discovery. Int J Mol Sci 2023; 24:3162. [PMID: 36834573 PMCID: PMC9963155 DOI: 10.3390/ijms24043162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/22/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Even with modern therapy, patients with heart failure only have a 50% five-year survival rate. To improve the development of new therapeutic strategies, preclinical models of disease are needed to properly emulate the human condition. Determining the most appropriate model represents the first key step for reliable and translatable experimental research. Rodent models of heart failure provide a strategic compromise between human in vivo similarity and the ability to perform a larger number of experiments and explore many therapeutic candidates. We herein review the currently available rodent models of heart failure, summarizing their physiopathological basis, the timeline of the development of ventricular failure, and their specific clinical features. In order to facilitate the future planning of investigations in the field of heart failure, a detailed overview of the advantages and possible drawbacks of each model is provided.
Collapse
Affiliation(s)
- Matteo Ponzoni
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Program in Translational Medicine, SickKids Research Institute, Toronto, ON M5G 0A4, Canada
| | - John G. Coles
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Program in Translational Medicine, SickKids Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jason T. Maynes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5G 1E2, Canada
| |
Collapse
|
14
|
Mustafa NH, Jalil J, Zainalabidin S, Saleh MS, Asmadi AY, Kamisah Y. Molecular mechanisms of sacubitril/valsartan in cardiac remodeling. Front Pharmacol 2022; 13:892460. [PMID: 36003518 PMCID: PMC9393311 DOI: 10.3389/fphar.2022.892460] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/11/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases have become a major clinical burden globally. Heart failure is one of the diseases that commonly emanates from progressive uncontrolled hypertension. This gives rise to the need for a new treatment for the disease. Sacubitril/valsartan is a new drug combination that has been approved for patients with heart failure. This review aims to detail the mechanism of action for sacubitril/valsartan in cardiac remodeling, a cellular and molecular process that occurs during the development of heart failure. Accumulating evidence has unveiled the cardioprotective effects of sacubitril/valsartan on cellular and molecular modulation in cardiac remodeling, with recent large-scale randomized clinical trials confirming its supremacy over other traditional heart failure treatments. However, its molecular mechanism of action in cardiac remodeling remains obscure. Therefore, comprehending the molecular mechanism of action of sacubitril/valsartan could help future research to study the drug's potential therapy to reduce the severity of heart failure.
Collapse
Affiliation(s)
- Nor Hidayah Mustafa
- Centre for Drug and Herbal Research Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Centre for Drug and Herbal Research Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Program of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohammed S.M. Saleh
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ahmad Yusof Asmadi
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur, Malaysia
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Nelson VL, Eadie AL, Brunt KR. A step towards a unifying pre-clinical model of dilated cardiomyopathy for drug development strategies or target validation. Am J Physiol Heart Circ Physiol 2022; 322:H1028-H1031. [PMID: 35427176 DOI: 10.1152/ajpheart.00141.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Victoria L Nelson
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, New Brunswick, Canada
| | - Ashley L Eadie
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, N.B., Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, NB, Canada
| |
Collapse
|
16
|
Spyropoulos F, Sorrentino A, van der Reest J, Yang P, Waldeck-Weiermair M, Steinhorn B, Eroglu E, Saeedi Saravi SS, Yu P, Haigis M, Christou H, Michel T. Metabolomic and transcriptomic signatures of chemogenetic heart failure. Am J Physiol Heart Circ Physiol 2022; 322:H451-H465. [PMID: 35089810 PMCID: PMC8896991 DOI: 10.1152/ajpheart.00628.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
The failing heart is characterized by elevated levels of reactive oxygen species. We have developed an animal model of heart failure induced by chemogenetic production of oxidative stress in the heart using a recombinant adeno-associated virus (AAV9) expressing yeast d-amino acid oxidase (DAAO) targeted to cardiac myocytes. When DAAO-infected animals are fed the DAAO substrate d-alanine, the enzyme generates hydrogen peroxide (H2O2) in the cardiac myocytes, leading to dilated cardiomyopathy. However, the underlying mechanisms of oxidative stress-induced heart failure remain incompletely understood. Therefore, we investigated the effects of chronic oxidative stress on the cardiac transcriptome and metabolome. Rats infected with recombinant cardiotropic AAV9 expressing DAAO or control AAV9 were treated for 7 wk with d-alanine to stimulate chemogenetic H2O2 production by DAAO and generate dilated cardiomyopathy. After hemodynamic assessment, left and right ventricular tissues were processed for RNA sequencing and metabolomic profiling. DAAO-induced dilated cardiomyopathy was characterized by marked changes in the cardiac transcriptome and metabolome both in the left and right ventricle. Downregulated transcripts are related to energy metabolism and mitochondrial function, accompanied by striking alterations in metabolites involved in cardiac energetics, redox homeostasis, and amino acid metabolism. Upregulated transcripts are involved in cytoskeletal organization and extracellular matrix. Finally, we noted increased metabolite levels of antioxidants glutathione and ascorbate. These findings provide evidence that chemogenetic generation of oxidative stress leads to a robust heart failure model with distinct transcriptomic and metabolomic signatures and set the basis for understanding the underlying pathophysiology of chronic oxidative stress in the heart.NEW & NOTEWORTHY We have developed a "chemogenetic" heart failure animal model that recapitulates a central feature of human heart failure: increased cardiac redox stress. We used a recombinant DAAO enzyme to generate H2O2 in cardiomyocytes, leading to cardiomyopathy. Here we report striking changes in the cardiac metabolome and transcriptome following chemogenetic heart failure, similar to changes observed in human heart failure. Our findings help validate chemogenetic approaches for the discovery of novel therapeutic targets in heart failure.
Collapse
Affiliation(s)
- Fotios Spyropoulos
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrea Sorrentino
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Peiran Yang
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Markus Waldeck-Weiermair
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin Steinhorn
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emrah Eroglu
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seyed Soheil Saeedi Saravi
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paul Yu
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marcia Haigis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
17
|
Chemogenetic approaches to dissect the role of H2O2 in redox-dependent pathways using genetically encoded biosensors. Biochem Soc Trans 2022; 50:335-345. [PMID: 35015078 DOI: 10.1042/bst20210506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
Chemogenetic tools are recombinant enzymes that can be targeted to specific organelles and tissues. The provision or removal of the enzyme substrate permits control of its biochemical activities. Yeast-derived enzyme D-amino acid oxidase (DAAO) represents the first of its kind for a substrate-based chemogenetic approach to modulate H2O2 concentrations within cells. Combining these powerful enzymes with multiparametric imaging methods exploiting genetically encoded biosensors has opened new lines of investigations in life sciences. In recent years, the chemogenetic DAAO approach has proven beneficial to establish a new role for (patho)physiological oxidative stress on redox-dependent signaling and metabolic pathways in cultured cells and animal model systems. This mini-review covers established or emerging methods and assesses newer approaches exploiting chemogenetic tools combined with genetically encoded biosensors.
Collapse
|
18
|
Steinhorn B, Eroglu E, Michel T. Chemogenetic Approaches to Probe Redox Pathways: Implications for Cardiovascular Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2022; 62:551-571. [PMID: 34530645 PMCID: PMC10507364 DOI: 10.1146/annurev-pharmtox-012221-082339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chemogenetics refers to experimental systems that dynamically regulate the activity of a recombinant protein by providing or withholding the protein's specific biochemical stimulus. Chemogenetic tools permit precise dynamic control of specific signaling molecules to delineate the roles of those molecules in physiology and disease. Yeast d-amino acid oxidase (DAAO) enables chemogenetic manipulation of intracellular redox balance by generating hydrogen peroxide only in the presence of d-amino acids. Advances in biosensors have allowed the precise quantitation of these signaling molecules. The combination of chemogenetic approaches with biosensor methodologies has opened up new lines of investigation, allowing the analysis of intracellular redox pathways that modulate physiological and pathological cell responses. We anticipate that newly developed transgenic chemogenetic models will permit dynamic modulation of cellularredox balance in diverse cells and tissues and will facilitate the identification and validation of novel therapeutic targets involved in both physiological redox pathways and pathological oxidative stress.
Collapse
Affiliation(s)
- Benjamin Steinhorn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Emrah Eroglu
- Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Department of Molecular Biology and Biochemistry, Medical University of Graz, 8036 Graz, Austria
| | - Thomas Michel
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
19
|
Erdogan YC, Altun HY, Secilmis M, Ata BN, Sevimli G, Cokluk Z, Zaki AG, Sezen S, Akgul Caglar T, Sevgen İ, Steinhorn B, Ai H, Öztürk G, Belousov VV, Michel T, Eroglu E. Complexities of the chemogenetic toolkit: Differential mDAAO activation by d-amino substrates and subcellular targeting. Free Radic Biol Med 2021; 177:132-142. [PMID: 34687864 PMCID: PMC8639799 DOI: 10.1016/j.freeradbiomed.2021.10.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023]
Abstract
A common approach to investigate oxidant-regulated intracellular pathways is to add exogenous H2O2 to living cells or tissues. However, the addition of H2O2 to the culture medium of cells or tissues approach does not accurately replicate intracellular redox-mediated cell responses. d-amino acid oxidase (DAAO)-based chemogenetic tools represent informative methodological advances that permit the generation of H2O2 on demand with a high spatiotemporal resolution by providing or withdrawing the DAAO substrate d-amino acids. Much has been learned about the intracellular transport of H2O2 through studies using DAAO, yet these valuable tools remain incompletely characterized in many cultured cells. In this study, we describe and characterize in detail the features of a new modified variant of DAAO (termed mDAAO) with improved catalytic activities. We tested mDAAO functionality in several cultured cell lines employing live-cell imaging techniques. Our imaging experiments show that mDAAO is suitable for the generation of H2O2 under hypoxic conditions imaged with the novel ultrasensitive H2O2 sensor (HyPer7). Moreover, this approach was suitable for generating H2O2 in a reversible and concentration-dependent manner in subcellular locales. Furthermore, we show that the choice of d-amino acids differentially affects mDAAO-dependent intracellular H2O2 generation. When paired with the hydrogen sulfide (H2S) sensor hsGFP, administration of the sulfur-containing amino acid d-cysteine to cells expressing mDAAO generates robust H2S signals. We also show that chemogenetic H2O2 generation in different cell types yields distinct HyPer7 profiles. These studies fully characterize the new mDAAO as a novel chemogenetic tool and provide multiparametric approaches for cell manipulation that may open new lines of investigations for redox biochemists to dissect the role of ROS signaling pathways with high spatial and temporal precision.
Collapse
Affiliation(s)
- Yusuf C Erdogan
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Hamza Y Altun
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Melike Secilmis
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Busra N Ata
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Gulsah Sevimli
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Zeynep Cokluk
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Asal Ghaffari Zaki
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Serap Sezen
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Tuba Akgul Caglar
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - İlker Sevgen
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Benjamin Steinhorn
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Gürkan Öztürk
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey; Physiology Department, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Vsevelod V Belousov
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Institute of Cardiovascular Physiology, Universitätsmedizin Göttingen, 37073, Göttingen, Germany
| | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Emrah Eroglu
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey; Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Austria; Nanotechnology Research and Application Center, Sabanci University, Istanbul, Turkey.
| |
Collapse
|
20
|
Waldeck-Weiermair M, Yadav S, Spyropoulos F, Krüger C, Pandey AK, Michel T. Dissecting in vivo and in vitro redox responses using chemogenetics. Free Radic Biol Med 2021; 177:360-369. [PMID: 34752919 PMCID: PMC8639655 DOI: 10.1016/j.freeradbiomed.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/08/2021] [Accepted: 11/04/2021] [Indexed: 02/03/2023]
Abstract
Hydrogen peroxide (H2O2) is the most abundant reactive oxygen species (ROS) within mammalian cells. At low concentrations, H2O2 serves as a versatile cell signaling molecule that mediates vital physiological functions. Yet at higher concentrations, H2O2 can be a toxic molecule by promoting pathological oxidative stress in cells and tissues. Within normal cells, H2O2 is differentially distributed in a variety of subcellular locales. Moreover, many redox-active enzymes and their substrates are themselves differentially distributed within cells. Numerous reports have described the biological and biochemical consequences of adding exogenous H2O2 to cultured cells and tissues, but many of these observations are difficult to interpret: the effects of exogenous H2O2 do not necessarily replicate the cellular responses to endogenous H2O2. In recent years, chemogenetic approaches have been developed to dynamically regulate the abundance of H2O2 in specific subcellular locales. Chemogenetic approaches have been applied in multiple experimental systems, ranging from in vitro studies on the intracellular transport and metabolism of H2O2, all the way to in vivo studies that generate oxidative stress in specific organs in living animals. These chemogenetic approaches have exploited a yeast-derived d-amino acid oxidase (DAAO) that synthesizes H2O2 only in the presence of its d-amino acid substrate. DAAO can be targeted to various subcellular locales, and can be dynamically activated by the addition or withdrawal of its d-amino acid substrate. In addition, recent advances in the development of highly sensitive genetically encoded H2O2 biosensors are providing a better understanding of both physiological and pathological oxidative pathways. This review highlights several applications of DAAO as a chemogenetic tool across a wide range of biological systems, from analyses of subcellular H2O2 metabolism in cells to the development of new disease models caused by oxidative stress in vivo.
Collapse
Affiliation(s)
- Markus Waldeck-Weiermair
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA; Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Shambhu Yadav
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Fotios Spyropoulos
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA; Department of Pediatric Newborn Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA
| | - Christina Krüger
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Differential Effects of Sacubitril/Valsartan on Diastolic Function in Mice With Obesity-Related Metabolic Heart Disease. JACC Basic Transl Sci 2020; 5:916-927. [PMID: 33015414 PMCID: PMC7524781 DOI: 10.1016/j.jacbts.2020.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
MHD associated with obesity, diabetes, and/or metabolic syndrome is an important precursor of HFpEF. Mice fed a HFHS diet develop MHD with myocardial hypertrophy, fibrosis, diastolic dysfunction, and impaired energetics. Mice on HFHS diet were treated with matched doses of VAL or sac SAC/VAL for 16 weeks. Only SAC/VAL prevented diastolic dysfunction and fibrosis, and to a lesser extent oxidative stress, whereas VAL and SAC/VAL had similar effects on hypertrophy and energetics. Neprilysin inhibition exerts beneficial effects on MHD that are complimentary to VAL, suggesting that SAC/VAL has promise to prevent the development of HFpEF in patients with or at risk for MHD.
Mice with obesity and metabolic heart disease (MHD) due to a high-fat, high-sucrose diet were treated with placebo, a clinically relevant dose of sacubitril (SAC)/valsartan (VAL), or an equivalent dose of VAL for 4 months. There were striking differences between SAC/VAL and VAL with regard to: 1) diastolic dysfunction; 2) interstitial fibrosis; and to a lesser degree; 3) oxidative stress—all of which were more favorably affected by SAC/VAL. SAC/VAL and VAL similarly attenuated myocardial hypertrophy and improved myocardial energetics. In mice with obesity-related MHD, neprilysin inhibition exerts favorable effects on diastolic function.
Collapse
Key Words
- ARB, angiotensin receptor blocker
- ATP, adenosine triphosphate
- CD, control diet
- GMP, guanosine monophosphate
- HFHS, high-fat, high-sucrose
- HFpEF, heart failure with a preserved ejection fraction
- LV, left ventricular
- MHD, metabolic heart disease
- MNA, methoxy-2-naphthlamine
- NMR, nuclear magnetic resonance
- PCr, phosphocreatine
- ROS, reactive oxygen species
- RPP, rate × pressure product
- SAC/VAL, sacubitril/valsartan
- VAL, valsartan
- diastolic
- neprilysin
- obesity
Collapse
|
22
|
Sorrentino A, Michel T. Redox à la carte: Novel chemogenetic models of heart failure. Br J Pharmacol 2020; 177:3162-3167. [PMID: 32368791 PMCID: PMC7312266 DOI: 10.1111/bph.15093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022] Open
Abstract
Many current animal models of heart failure are hampered by intrinsic methodological complexities, while other models yield only a subtle cardiac phenotype even after prolonged in vivo treatments. A new 'chemogenetic' animal model of heart failure reproduces a critical characteristic shared by many disease states that lead to heart failure in humans: an increase in redox stress in the heart. This 'chemogenetic' approach exploits a recombinant yeast enzyme that can be dynamically and specifically activated in vivo to generate the ROS hydrogen peroxide (H2 O2 ) in cardiac myocytes. Redox stress can be rapidly, selectively and reversibly manipulated by chemogenetic generation of ROS in cardiac myocytes, yielding a new model of dilated cardiomyopathy. Treatment of animals with the angiotensin receptor antagonist valsartan promotes recovery of ventricular function and resolution of adverse cardiac remodelling. This mini-review discusses in vivo chemogenetic approaches to manipulate and analyse oxidative stress in the heart.
Collapse
Affiliation(s)
- Andrea Sorrentino
- Cardiovascular Division, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Thomas Michel
- Cardiovascular Division, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|