51
|
Li Q, Zhang S, Yang G, Wang X, Liu F, Li Y, Chen Y, Zhou T, Xie D, Liu Y, Zhang L. Energy metabolism: A critical target of cardiovascular injury. Biomed Pharmacother 2023; 165:115271. [PMID: 37544284 DOI: 10.1016/j.biopha.2023.115271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Cardiovascular diseases are the main killers threatening human health. Many studies have shown that abnormal energy metabolism plays a key role in the occurrence and development of acute and chronic cardiovascular diseases. Regulating cardiac energy metabolism is a frontier topic in the treatment of cardiovascular diseases. However, we are not very clear about the choice of different substrates, the specific mechanism of energy metabolism participating in the course of cardiovascular disease, and how to develop appropriate drugs to regulate energy metabolism to treat cardiovascular disease. Therefore, this paper reviews how energy metabolism participates in cardiovascular pathophysiological processes and potential drugs aimed at interfering energy metabolism.It is expected to provide good suggestions for promoting the clinical prevention and treatment of cardiovascular diseases from the perspective of energy metabolism.
Collapse
Affiliation(s)
- Qiyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gengqiang Yang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Fuxian Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Dingxiong Xie
- Gansu Institute of Cardiovascular Diseases, LanZhou, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation Ministry of Education, China.
| | - Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Gansu Institute of Cardiovascular Diseases, LanZhou, China.
| |
Collapse
|
52
|
Prag HA, Murphy MP, Krieg T. Preventing mitochondrial reverse electron transport as a strategy for cardioprotection. Basic Res Cardiol 2023; 118:34. [PMID: 37639068 PMCID: PMC10462584 DOI: 10.1007/s00395-023-01002-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
In the context of myocardial infarction, the burst of superoxide generated by reverse electron transport (RET) at complex I in mitochondria is a crucial trigger for damage during ischaemia/reperfusion (I/R) injury. Here we outline the necessary conditions for superoxide production by RET at complex I and how it can occur during reperfusion. In addition, we explore various pathways that are implicated in generating the conditions for RET to occur and suggest potential therapeutic strategies to target RET, aiming to achieve cardioprotection.
Collapse
Affiliation(s)
- Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
53
|
Bodnar P, Mazurkiewicz M, Chwalba T, Romuk E, Ciszek-Chwalba A, Jacheć W, Wojciechowska C. The Impact of Pharmacotherapy for Heart Failure on Oxidative Stress-Role of New Drugs, Flozins. Biomedicines 2023; 11:2236. [PMID: 37626732 PMCID: PMC10452694 DOI: 10.3390/biomedicines11082236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Heart failure (HF) is a multifactorial clinical syndrome involving many complex processes. The causes may be related to abnormal heart structure and/or function. Changes in the renin-angiotensin-aldosterone system, the sympathetic nervous system, and the natriuretic peptide system are important in the pathophysiology of HF. Dysregulation or overexpression of these processes leads to changes in cardiac preload and afterload, changes in the vascular system, peripheral vascular dysfunction and remodeling, and endothelial dysfunction. One of the important factors responsible for the development of heart failure at the cellular level is oxidative stress. This condition leads to deleterious cellular effects as increased levels of free radicals gradually disrupt the state of equilibrium, and, as a consequence, the internal antioxidant defense system is damaged. This review focuses on pharmacotherapy for chronic heart failure with regard to oxidation-reduction metabolism, with special attention paid to the latest group of drugs, SGLT2 inhibitors-an integral part of HF treatment. These drugs have been shown to have beneficial effects by protecting the antioxidant system at the cellular level.
Collapse
Affiliation(s)
- Patryk Bodnar
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (P.B.); (T.C.); (A.C.-C.)
| | | | - Tomasz Chwalba
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (P.B.); (T.C.); (A.C.-C.)
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Street, 41-808 Zabrze, Poland
| | - Anna Ciszek-Chwalba
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (P.B.); (T.C.); (A.C.-C.)
| | - Wojciech Jacheć
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.J.); (C.W.)
| | - Celina Wojciechowska
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.J.); (C.W.)
| |
Collapse
|
54
|
Iuchi K, Fukasawa M, Murakami T, Hisatomi H. Cold atmospheric nitrogen plasma induces metal-initiated cell death by cell membrane rupture and mitochondrial perturbation. Cell Biochem Funct 2023; 41:687-695. [PMID: 37322606 DOI: 10.1002/cbf.3823] [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: 11/05/2022] [Revised: 05/06/2023] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Cold atmospheric plasma (CAP) is a novel biomedical tool used for cancer therapy. A device using nitrogen gas (N2 CAP) produced CAP that induced cell death through the production of reactive nitrogen species and an increase in intracellular calcium. In this study, we investigated the effect of N2 CAP-irradiation on cell membrane and mitochondrial function in human embryonic kidney cell line 293T. We investigated whether iron is involved in N2 CAP-induced cell death, as deferoxamine methanesulfonate (an iron chelator) inhibits this process. We found that N2 CAP induced cell membrane disturbance and loss of mitochondrial membrane potential in an irradiation time-dependent manner. BAPTA-AM, a cell-permeable calcium chelator, inhibited N2 CAP-induced loss of mitochondrial membrane potential. These results suggest that disruption of intracellular metal homeostasis was involved in N2 CAP-induced cell membrane rupture and mitochondrial dysfunction. Moreover, N2 CAP irradiation generated a time-dependent production of peroxynitrite. However, lipid-derived radicals are unrelated to N2 CAP-induced cell death. Generally, N2 CAP-induced cell death is driven by the complex interaction between metal movement and reactive oxygen and nitrogen species produced by N2 CAP.
Collapse
Affiliation(s)
- Katsuya Iuchi
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Tokyo, Japan
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama, Japan
| | - Mami Fukasawa
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Tokyo, Japan
| | - Tomoyuki Murakami
- Department of Systems Design Engineering, Faculty of Science and Technology, Seikei University, Tokyo, Japan
| | - Hisashi Hisatomi
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Tokyo, Japan
| |
Collapse
|
55
|
Algoet M, Janssens S, Himmelreich U, Gsell W, Pusovnik M, Van den Eynde J, Oosterlinck W. Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends Cardiovasc Med 2023; 33:357-366. [PMID: 35181472 DOI: 10.1016/j.tcm.2022.02.005] [Citation(s) in RCA: 191] [Impact Index Per Article: 95.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/20/2022]
Abstract
Acute myocardial infarction is caused by a sudden coronary artery occlusion and leads to ischemia in the corresponding myocardial territory which generally results in myocardial necrosis. Without restoration of coronary perfusion, myocardial scar formation will cause adverse remodelling of the myocardium and heart failure. Successful introduction of percutaneous coronary intervention and surgical coronary artery bypass grafting made it possible to achieve early revascularisation/reperfusion, hence limiting the ischemic zone of myocardium. However, reperfusion by itself paradoxically triggers an exacerbated and accelerated injury in the myocardium, called ischemia-reperfusion (I/R) injury. This mechanism is partially driven by inflammation through multiple interacting pathways. In this review we summarize the current insights in mechanisms of I/R injury and the influence of altered inflammation. Multiple pharmacological and interventional therapeutic strategies (ischemic conditioning) have proven to be beneficial during I/R in preclinical models but were notoriously unsuccessful upon clinical translation. In this review we focus on common mechanisms of I/R injury, altered inflammation and potential therapeutic strategies. We hypothesize that a dual approach may be of value because I/R injury patients are predestined with multiple comorbidities and systemic low-grade inflammation, which requires targeted intervention before other strategies can be fully effective.
Collapse
Affiliation(s)
- Michiel Algoet
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Matic Pusovnik
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jef Van den Eynde
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Helen B. Taussig Heart Center, The Johns Hopkins Hospital and School of Medicine, Baltimore, United States
| | | |
Collapse
|
56
|
Ponnusamy T, Velusamy P, Kumar A, Morris D, Zhang X, Ning G, Klinger M, Copper JE, Rajan S, Cheung JY, Natarajaseenivasan K, Mnatsakanyan N, Shanmughapriya S. Mitochondrial Magnesium is the cationic rheostat for MCU-mediated mitochondrial Ca 2+ uptake. RESEARCH SQUARE 2023:rs.3.rs-3088175. [PMID: 37502932 PMCID: PMC10371168 DOI: 10.21203/rs.3.rs-3088175/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Calcium (Ca2+) uptake by mitochondria is essential in regulating bioenergetics, cell death, and cytosolic Ca2+ transients. Mitochondrial Calcium Uniporter (MCU) mediates the mitochondrial Ca2+ uptake. MCU is a heterooligomeric complex with a pore-forming component and accessory proteins required for channel activity. Though MCU regulation by MICUs is unequivocally established, there needs to be more knowledge of whether divalent cations regulate MCU. Here we set out to understand the mitochondrial matrix Mg2+-dependent regulation of MCU activity. We showed Mrs2 as the authentic mammalian mitochondrial Mg2+ channel using the planar lipid bilayer recordings. Using a liver-specific Mrs2 KO mouse model, we showed that decreased matrix [Mg2+] is associated with increased MCU activity and matrix Ca2+ overload. The disruption of Mg2+dependent MCU regulation significantly prompted mitochondrial permeability transition pore opening-mediated cell death during tissue IR injury. Our findings support a critical role for mMg2+ in regulating MCU activity and attenuating mCa2+ overload.
Collapse
Affiliation(s)
- Thiruvelselvan Ponnusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Amrendra Kumar
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Daniel Morris
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Xueqian Zhang
- Cardiovascular Medicine, Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Gang Ning
- Microscopy Core Facility, Penn State Huck Institutes of the Life Sciences, University Park, PA 16802, USA
| | - Marianne Klinger
- Department of Pathology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Jean E. Copper
- Department of Pathology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Sudarsan Rajan
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Joseph Y Cheung
- Department of Renal Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Nelli Mnatsakanyan
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
57
|
She R, Liu D, Liao J, Wang G, Ge J, Mei Z. Mitochondrial dysfunctions induce PANoptosis and ferroptosis in cerebral ischemia/reperfusion injury: from pathology to therapeutic potential. Front Cell Neurosci 2023; 17:1191629. [PMID: 37293623 PMCID: PMC10244524 DOI: 10.3389/fncel.2023.1191629] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
Ischemic stroke (IS) accounts for more than 80% of the total stroke, which represents the leading cause of mortality and disability worldwide. Cerebral ischemia/reperfusion injury (CI/RI) is a cascade of pathophysiological events following the restoration of blood flow and reoxygenation, which not only directly damages brain tissue, but also enhances a series of pathological signaling cascades, contributing to inflammation, further aggravate the damage of brain tissue. Paradoxically, there are still no effective methods to prevent CI/RI, since the detailed underlying mechanisms remain vague. Mitochondrial dysfunctions, which are characterized by mitochondrial oxidative stress, Ca2+ overload, iron dyshomeostasis, mitochondrial DNA (mtDNA) defects and mitochondrial quality control (MQC) disruption, are closely relevant to the pathological process of CI/RI. There is increasing evidence that mitochondrial dysfunctions play vital roles in the regulation of programmed cell deaths (PCDs) such as ferroptosis and PANoptosis, a newly proposed conception of cell deaths characterized by a unique form of innate immune inflammatory cell death that regulated by multifaceted PANoptosome complexes. In the present review, we highlight the mechanisms underlying mitochondrial dysfunctions and how this key event contributes to inflammatory response as well as cell death modes during CI/RI. Neuroprotective agents targeting mitochondrial dysfunctions may serve as a promising treatment strategy to alleviate serious secondary brain injuries. A comprehensive insight into mitochondrial dysfunctions-mediated PCDs can help provide more effective strategies to guide therapies of CI/RI in IS.
Collapse
Affiliation(s)
- Ruining She
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Danhong Liu
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jun Liao
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
58
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
59
|
Wang H, Liu D, Zheng B, Yang Y, Qiao Y, Li S, Pan S, Liu Y, Feng Q, Liu Z. Emerging Role of Ferroptosis in Diabetic Kidney Disease: Molecular Mechanisms and Therapeutic Opportunities. Int J Biol Sci 2023; 19:2678-2694. [PMID: 37324941 PMCID: PMC10266077 DOI: 10.7150/ijbs.81892] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common and severe microvascular complications of diabetes mellitus (DM), and has become the leading cause of end-stage renal disease (ESRD) worldwide. Although the exact pathogenic mechanism of DKD is still unclear, programmed cell death has been demonstrated to participate in the occurrence and development of diabetic kidney injury, including ferroptosis. Ferroptosis, an iron-dependent form of cell death driven by lipid peroxidation, has been identified to play a vital role in the development and therapeutic responses of a variety of kidney diseases, such as acute kidney injury (AKI), renal cell carcinoma and DKD. In the past two years, ferroptosis has been well investigated in DKD patients and animal models, but the specific mechanisms and therapeutic effects have not been fully revealed. Herein, we reviewed the regulatory mechanisms of ferroptosis, summarized the recent findings associated with the involvement of ferroptosis in DKD, and discussed the potential of ferroptosis as a promising target for DKD treatment, thereby providing a valuable reference for basic study and clinical therapy of DKD.
Collapse
Affiliation(s)
- Hui Wang
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Dongwei Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Bin Zheng
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Yang Yang
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Yingjin Qiao
- Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Shiyang Li
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Shaokang Pan
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Yong Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| | - Zhangsuo Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, P. R. China
| |
Collapse
|
60
|
Wang B, Wang Y, Zhang J, Hu C, Jiang J, Li Y, Peng Z. ROS-induced lipid peroxidation modulates cell death outcome: mechanisms behind apoptosis, autophagy, and ferroptosis. Arch Toxicol 2023; 97:1439-1451. [PMID: 37127681 DOI: 10.1007/s00204-023-03476-6] [Citation(s) in RCA: 199] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/02/2023] [Indexed: 05/03/2023]
Abstract
Reactive oxygen species (ROS) mediate lipid peroxidation and produce 4-hydroxynonenal and other related products, which play an important role in the process of cell death, including apoptosis, autophagy, and ferroptosis. Lipid peroxidation of phospholipid bilayers can promote mitochondrial apoptosis, endoplasmic reticulum stress, and other complex molecular signaling pathways to regulate apoptosis. Lipid peroxidation and its products also act at different stages of autophagy, affecting the formation of autophagosomes and the recruitment of downstream proteins. In addition, we discuss the important role of ROS and lipid peroxides in ferroptosis and the regulatory role of nuclear factor erythroid 2-related factor 2 in ferroptosis under a background of oxidation. Finally, from the perspectives of promotion, inhibition, transformation, and common upstream molecules, we summarized the crosstalk among apoptosis, autophagy, and ferroptosis in the context of ROS. Our review discusses the role of ROS and lipid peroxidation in apoptosis, autophagy, and ferroptosis and their possible crosstalk mechanisms, so as to provide new insights and directions for the study of diseases related to pathological cell death. This review also has referential significance for studying the exact mechanism of ferroptosis mediated by lipid peroxidation.
Collapse
Affiliation(s)
- Bingqing Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yue Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Chang Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Jun Jiang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - ZhiYong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
- Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
61
|
Murakami Y, Wei FY, Kawamura Y, Horiguchi H, Kadomatsu T, Miyata K, Miura K, Oike Y, Ando Y, Ueda M, Tomizawa K, Chujo T. NSUN3-mediated mitochondrial tRNA 5-formylcytidine modification is essential for embryonic development and respiratory complexes in mice. Commun Biol 2023; 6:307. [PMID: 36949224 PMCID: PMC10033821 DOI: 10.1038/s42003-023-04680-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2023] [Indexed: 03/24/2023] Open
Abstract
In mammalian mitochondria, translation of the AUA codon is supported by 5-formylcytidine (f5C) modification in the mitochondrial methionine tRNA anticodon. The 5-formylation is initiated by NSUN3 methylase. Human NSUN3 mutations are associated with mitochondrial diseases. Here we show that Nsun3 is essential for embryonic development in mice with whole-body Nsun3 knockout embryos dying between E10.5 and E12.5. To determine the functions of NSUN3 in adult tissue, we generated heart-specific Nsun3 knockout (Nsun3HKO) mice. Nsun3HKO heart mitochondria were enlarged and contained fragmented cristae. Nsun3HKO resulted in enhanced heart contraction and age-associated mild heart enlargement. In the Nsun3HKO hearts, mitochondrial mRNAs that encode respiratory complex subunits were not down regulated, but the enzymatic activities of the respiratory complexes decreased, especially in older mice. Our study emphasizes that mitochondrial tRNA anticodon modification is essential for mammalian embryonic development and shows that tissue-specific loss of a single mitochondrial tRNA modification can induce tissue aberration that worsens in later adulthood.
Collapse
Affiliation(s)
- Yoshitaka Murakami
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Neurology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Fan-Yan Wei
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan
| | - Yoshimi Kawamura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kyoko Miura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yukio Ando
- Department of Amyloidosis Research, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, 859-3298, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Takeshi Chujo
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
62
|
Olanlokun JO, Oyebode OT, Popoola D, Bodede O, Idowu TO, Moodley R, Olorunsogo OO. In vitro effects of 2-methyl-3-propylbutane-1,4-diol purified from Alstonia boonei on erythrocyte membrane stabilization and mitochondrial membrane permeabilization. Chem Biol Drug Des 2023; 101:678-689. [PMID: 36323654 DOI: 10.1111/cbdd.14168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022]
Abstract
A recent review on the ethnomedicinal, chemical, pharmacological, and toxicological properties of Alstonia boonei revealed the plant's potential in the treatment and management of a range of diseases. However, most of these pharmacological effects are only traceable to the crude form of the plant extract and not specific natural products. Phytochemical investigation of the methanol fraction of the methanol extract of the stem-bark of Alstonia boonei led to the isolation and identification of 2-methyl-3-propylbutane-1,4-diol. The structures were elucidated by the application of 1D-, and 2D-NMR spectroscopic analyses and by comparison with literature data. In this study, the membrane stabilizing activity, mitochondrial membrane permeability transition pore opening, cytochrome c release, mitochondrial ATPase activity, and prevention of mitochondrial lipid peroxidation activity of 2-methyl-3-propylbutane-1,4-diol (MPBD) isolated from A. boonei were determined. The results showed that MPBD significantly (p < .05) prevented peroxidation of mitochondrial membrane lipids and hemolysis using both the heat-induced and hypotonic solution-induced membrane stabilization assays. On the contrary, the compound caused large amplitude swelling of rat liver mitochondria in the absence of calcium, significant (p < .05) cytochrome c release and enhancement of mitochondrial ATPase activity in vitro. Our findings suggest that MPBD showed characteristic biological properties useful in modulating cell death.
Collapse
Affiliation(s)
- John Oludele Olanlokun
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olubukola Titilope Oyebode
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - David Popoola
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olusola Bodede
- Natural Products Research Laboratory, School of Chemistry and Physics, University of Kwazulu-Natal, Durban, South Africa.,Department of Chemistry, University of Pretoria, Pretoria, South Africa
| | - Thomas Oyebode Idowu
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Roshila Moodley
- Natural Products Research Laboratory, School of Chemistry and Physics, University of Kwazulu-Natal, Durban, South Africa
| | - Olufunso Olabode Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
63
|
Calcium signaling and genetic rare diseases: An auditory perspective. Cell Calcium 2023; 110:102702. [PMID: 36791536 DOI: 10.1016/j.ceca.2023.102702] [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: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Deafness is a highly heterogeneous disorder which stems, for 50%, from genetic origins. Sensory transduction relies mainly on sensory hair cells of the cochlea, in the inner ear. Calcium is key for the function of these cells and acts as a fundamental signal transduction. Its homeostasis depends on three factors: the calcium influx, through the mechanotransduction channel at the apical pole of the hair cell as well as the voltage-gated calcium channel at the base of the cells; the calcium buffering via Ca2+-binding proteins in the cytoplasm, but also in organelles such as mitochondria and the reticulum endoplasmic mitochondria-associated membranes with specialized proteins; and the calcium extrusion through the Ca-ATPase pump, located all over the plasma membrane. In addition, the synaptic transmission to the central nervous system is also controlled by calcium. Genetic studies of inherited deafness have tremendously helped understand the underlying molecular pathways of calcium signaling. In this review, we discuss these different factors in light of the associated genetic diseases (syndromic and non-syndromic deafness) and the causative genes.
Collapse
|
64
|
Galeone A, Grano M, Brunetti G. Tumor Necrosis Factor Family Members and Myocardial Ischemia-Reperfusion Injury: State of the Art and Therapeutic Implications. Int J Mol Sci 2023; 24:4606. [PMID: 36902036 PMCID: PMC10003149 DOI: 10.3390/ijms24054606] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Ischemic heart disease is the principal cause of death worldwide and clinically manifests as myocardial infarction (MI), stable angina, and ischemic cardiomyopathy. Myocardial infarction is defined as an irreversible injury due to severe and prolonged myocardial ischemia inducing myocardial cell death. Revascularization is helpful in reducing loss of contractile myocardium and improving clinical outcome. Reperfusion rescues myocardium from cell death but also induces an additional injury called ischemia-reperfusion injury. Multiple mechanisms are involved in ischemia-reperfusion injury, such as oxidative stress, intracellular calcium overload, apoptosis, necroptosis, pyroptosis, and inflammation. Various members of the tumor necrosis factor family play a key role in myocardial ischemia-reperfusion injury. In this article, the role of TNFα, CD95L/CD95, TRAIL, and the RANK/RANKL/OPG axis in the regulation of myocardial tissue damage is reviewed together with their potential use as a therapeutic target.
Collapse
Affiliation(s)
- Antonella Galeone
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| |
Collapse
|
65
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
66
|
Wu H, Liu Y, Hao Y, Hou D, Yang R. Lycium barbarum polysaccharide protects cardiomyocytes from hypoxia/reoxygenation injury via activation of SIRT3/CypD signaling. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:72. [PMID: 36819526 PMCID: PMC9929766 DOI: 10.21037/atm-22-6081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/03/2023] [Indexed: 02/03/2023]
Abstract
Background Myocardial ischemia-reperfusion is a common pathological feature of many heart and vascular diseases, but the molecular mechanism of this process is still unclear, and there is no effective way to protect cardiomyocytes. The aim of this study was to examine the effects and underlying molecular mechanisms of Lycium barbarum polysaccharide (LBP) on myocardial ischemia-reperfusion injury in cardiomyocytes. Methods The cardiomyocyte cell line H9c2 were used to establish an in vitro hypoxia/reoxygenation (H/R) model. After treatment with LBP and/or the SIRT3 inhibitor 3-TYP, cell morphology was observed under the light microscopy. The Cell Counting Kit (CCK)-8 and 5-ethynyl-2'-deoxyuridine (EdU) assay were used to detect cell proliferation, and flow cytometry was performed to assess cell apoptosis. The lysine (166)-acetylation of CypD1 was determined by co-immunoprecipitation assay. Enzyme-linked immunosorbent assay (ELISA) was used to determine the lactate dehydrogenase (LDH) level in the culture medium. Na+-K+-ATPase activity, Ca2+-ATPase activity, and nitric oxide (NO) levels were measured. Results LBP alleviated cell damage and upregulated STIR3 expression in a dose-dependent manner. Upregulated SIRT3 expression and suppressed acetylation of CypD were also observed in H/R-induced H9c2 cells treated with LBP. Indeed, LBP remarkably reversed the inhibition of proliferation and cell apoptosis in H/R-induced H9c2 cells by activating SIRT3/CypD signaling. Blockade of SIRT3 with SIRT3 inhibitor (3-TYP) inhibited the protective effect of LBP on H9c2 cells. LBP markedly alleviated the H/R-induced increase of LDH release, and the decrease of Na+-K+-ATPase activity, Ca2+-ATPase activity, and NO levels. Inhibition of SIRT3 restored the protective effects of LBP. Conclusions LPB induced deacetylation of CypD by upregulating SIRT3, thereby protecting mitochondrial function and relieving H/R-induced injury in cardiomyocytes.
Collapse
Affiliation(s)
- Hailiang Wu
- Cadre Ward of Heart Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yajuan Liu
- Cadre Ward of Heart Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yu Hao
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Dandan Hou
- Cadre Ward of Heart Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ruiying Yang
- Cadre Ward of Heart Center, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
67
|
Makievskaya CI, Popkov VA, Andrianova NV, Liao X, Zorov DB, Plotnikov EY. Ketogenic Diet and Ketone Bodies against Ischemic Injury: Targets, Mechanisms, and Therapeutic Potential. Int J Mol Sci 2023; 24:2576. [PMID: 36768899 PMCID: PMC9916612 DOI: 10.3390/ijms24032576] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
The ketogenic diet (KD) has been used as a treatment for epilepsy since the 1920s, and its role in the prevention of many other diseases is now being considered. In recent years, there has been an intensive investigation on using the KD as a therapeutic approach to treat acute pathologies, including ischemic ones. However, contradictory data are observed for the effects of the KD on various organs after ischemic injury. In this review, we provide the first systematic analysis of studies conducted from 1980 to 2022 investigating the effects and main mechanisms of the KD and its mimetics on ischemia-reperfusion injury of the brain, heart, kidneys, liver, gut, and eyes. Our analysis demonstrated a high diversity of both the composition of the used KD and the protocols for the treatment of animals, which could be the reason for contradictory effects in different studies. It can be concluded that a true KD or its mimetics, such as β-hydroxybutyrate, can be considered as positive exposure, protecting the organ from ischemia and its negative consequences, whereas the shift to a rather similar high-calorie or high-fat diet leads to the opposite effect.
Collapse
Affiliation(s)
- Ciara I. Makievskaya
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Xinyu Liao
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| |
Collapse
|
68
|
Mitochondrial remodelling is essential for female germ cell differentiation and survival. PLoS Genet 2023; 19:e1010610. [PMID: 36696418 PMCID: PMC9901744 DOI: 10.1371/journal.pgen.1010610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/06/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Stem cells often possess immature mitochondria with few inner membrane invaginations, which increase as stem cells differentiate. Despite this being a conserved feature across many stem cell types in numerous organisms, how and why mitochondria undergo such remodelling during stem cell differentiation has remained unclear. Here, using Drosophila germline stem cells (GSCs), we show that Complex V drives mitochondrial remodelling during the early stages of GSC differentiation, prior to terminal differentiation. This endows germline mitochondria with the capacity to generate large amounts of ATP required for later egg growth and development. Interestingly, impairing mitochondrial remodelling prior to terminal differentiation results in endoplasmic reticulum (ER) lipid bilayer stress, Protein kinase R-like ER kinase (PERK)-mediated activation of the Integrated Stress Response (ISR) and germ cell death. Taken together, our data suggest that mitochondrial remodelling is an essential and tightly integrated aspect of stem cell differentiation. This work sheds light on the potential impact of mitochondrial dysfunction on stem and germ cell function, highlighting ER lipid bilayer stress as a potential major driver of phenotypes caused by mitochondrial dysfunction.
Collapse
|
69
|
Reyes Gaido OE, Schole KL, Anderson ME, Luczak ED. Genome-wide CRISPR screen reveals genetic modifiers of Ca 2+ -mediated cell death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523980. [PMID: 36712017 PMCID: PMC9882248 DOI: 10.1101/2023.01.13.523980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Ca 2+ is a fundamental determinant of survival in living cells. Excessive intracellular Ca 2+ causes cellular toxicity and death but the genetic pathways contributing to Ca 2+ induced cell death are incompletely understood. Here, we performed genome-wide CRISPR knock-out screening in human cells challenged with the Ca 2+ ionophore ionomycin and identified genes and pathways essential for cell death after Ca 2+ overload. We discovered 115 protective gene knockouts, 82 of which are non-essential genes and 21 of which belong to the druggable genome. Notably, members of store operated Ca 2+ entry (SOCE), very long-chain fatty acid synthesis, and SWItch/Sucrose Non-Fermentable (SWI/SNF) pathways provided marked protection against Ca 2+ toxicity. These results reveal pathways previously unknown to mediate Ca 2+ -induced cell death and provide a resource for the development of pharmacotherapies against the sequelae of Ca 2+ overload in disease.
Collapse
|
70
|
Xing R, Cheng J, Yu J, Li S, Ma H, Zhao Y. Trifluoperazine reduces apoptosis and inflammatory responses in traumatic brain injury by preventing the accumulation of Aquaporin4 on the surface of brain cells. Int J Med Sci 2023; 20:797-809. [PMID: 37213674 PMCID: PMC10198142 DOI: 10.7150/ijms.82677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/07/2023] [Indexed: 05/23/2023] Open
Abstract
Currently, no specific and standard treatment for traumatic brain injury (TBI) has been developed. Therefore, studies on new therapeutic drugs for TBI treatment are urgently needed. Trifluoperazine (TFP) is a therapeutic agent for the treatment of psychiatric disorders that reduces edema of the central nervous system. However, the specific working mechanism of TFP is not fully understood in TBI. In this study, the immunofluorescence co-localization analysis revealed that the area and intensity covered by Aquaporin4 (AQP4) on the surface of brain cells (astrocyte endfeet) increased significantly after TBI. In contrast, TFP treatment reversed these phenomena. This finding showed that TFP inhibited AQP4 accumulation on the surface of brain cells (astrocyte endfeet). The tunel fluorescence intensity and fluorescence area were lower in the TBI+TFP group compared to the TBI group. Additionally, the brain edema, brain defect area, and modified neurological severity score (mNSS) were lower in the TBI+TFP. The RNA-seq was performed on the cortical tissues of rats in the Sham, TBI, and TBI+TFP groups. A total of 3774 genes differently expressed between the TBI and the Sham group were identified. Of these, 2940 genes were up-regulated and 834 genes were down-regulated. A total of 1845 differently expressed genes between the TBI+TFP and TBI group were also identified, in which 621 genes were up-regulated and 1224 genes were down-regulated. Analysis of the common differential genes in the three groups showed that TFP could reverse the expression of apoptosis and inflammation genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that the differentially expressed genes (DEGs) were highly enriched in the signaling pathways regulating inflammation. In conclusion, TFP alleviates brain edema after TBI by preventing the accumulation of AQP4 on the surface of brain cells. Generally, TFP alleviates apoptosis and inflammatory response induced by TBI, and promotes the recovery of nerve function in rats after TBI. Thus, TFP is a potential therapeutic agent for TBI treatment.
Collapse
Affiliation(s)
- Rongchun Xing
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jin Cheng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shaoping Li
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haoli Ma
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- ✉ Corresponding authors: Haoli Ma, ; Yan Zhao,
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- ✉ Corresponding authors: Haoli Ma, ; Yan Zhao,
| |
Collapse
|
71
|
Algieri C, Bernardini C, Marchi S, Forte M, Tallarida MA, Bianchi F, La Mantia D, Algieri V, Stanzione R, Cotugno M, Costanzo P, Trombetti F, Maiuolo L, Forni M, De Nino A, Di Nonno F, Sciarretta S, Volpe M, Rubattu S, Nesci S. 1,5-disubstituted-1,2,3-triazoles counteract mitochondrial dysfunction acting on F 1F O-ATPase in models of cardiovascular diseases. Pharmacol Res 2023; 187:106561. [PMID: 36410676 DOI: 10.1016/j.phrs.2022.106561] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
The compromised viability and function of cardiovascular cells are rescued by small molecules of triazole derivatives (Tzs), identified as 3a and 3b, by preventing mitochondrial dysfunction. The oxidative phosphorylation improves the respiratory control rate in the presence of Tzs independently of the substrates that energize the mitochondria. The F1FO-ATPase, the main candidate in mitochondrial permeability transition pore (mPTP) formation, is the biological target of Tzs and hydrophilic F1 domain of the enzyme is depicted as the binding region of Tzs. The protective effect of Tz molecules on isolated mitochondria was corroborated by immortalized cardiomyocytes results. Indeed, mPTP opening was attenuated in response to ionomycin. Consequently, increased mitochondrial roundness and reduction of both length and interconnections between mitochondria. In in-vitro and ex-vivo models of cardiovascular pathologies (i.e., hypoxia-reoxygenation and hypertension) were used to evaluate the Tzs cardioprotective action. Key parameters of porcine aortic endothelial cells (pAECs) oxidative metabolism and cell viability were not affected by Tzs. However, in the presence of either 1 μM 3a or 0.5 μM 3b the impaired cell metabolism of pAECs injured by hypoxia-reoxygenation was restored to control respiratory profile. Moreover, endothelial cells isolated from SHRSP exposed to high-salt treatment rescued the Complex I activity and the endothelial capability to form vessel-like tubes and vascular function in presence of Tzs. As a result, the specific biochemical mechanism of Tzs to block Ca2+-activated F1FO-ATPase protected cell viability and preserved the pAECs bioenergetic metabolism upon hypoxia-reoxygenation injury. Moreover, SHRSP improved vascular dysfunction in response to a high-salt treatment.
Collapse
Affiliation(s)
- Cristina Algieri
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona 60126, Italy
| | | | | | | | - Debora La Mantia
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy
| | - Vincenzo Algieri
- Department of Chemistry and Chemical Technologies, University of Calabria, Cosenza 87036, Italy
| | | | | | - Paola Costanzo
- Department of Chemistry and Chemical Technologies, University of Calabria, Cosenza 87036, Italy
| | - Fabiana Trombetti
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy
| | - Loredana Maiuolo
- Department of Chemistry and Chemical Technologies, University of Calabria, Cosenza 87036, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy; Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), Alma Mater Studiorum-University of Bologna, Bologna 40126, Italy
| | - Antonio De Nino
- Department of Chemistry and Chemical Technologies, University of Calabria, Cosenza 87036, Italy
| | | | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli 86077, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina 04100, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome 00189, Italy; IRCCS San Raffaele, Rome 00163, Italy
| | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli 86077, Italy; Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome 00189, Italy
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia 40064, Italy.
| |
Collapse
|
72
|
Huang X, Zeng Z, Li S, Xie Y, Tong X. The Therapeutic Strategies Targeting Mitochondrial Metabolism in Cardiovascular Disease. Pharmaceutics 2022; 14:pharmaceutics14122760. [PMID: 36559254 PMCID: PMC9788260 DOI: 10.3390/pharmaceutics14122760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is a group of systemic disorders threatening human health with complex pathogenesis, among which mitochondrial energy metabolism reprogramming has a critical role. Mitochondria are cell organelles that fuel the energy essential for biochemical reactions and maintain normal physiological functions of the body. Mitochondrial metabolic disorders are extensively involved in the progression of CVD, especially for energy-demanding organs such as the heart. Therefore, elucidating the role of mitochondrial metabolism in the progression of CVD is of great significance to further understand the pathogenesis of CVD and explore preventive and therapeutic methods. In this review, we discuss the major factors of mitochondrial metabolism and their potential roles in the prevention and treatment of CVD. The current application of mitochondria-targeted therapeutic agents in the treatment of CVD and advances in mitochondria-targeted gene therapy technologies are also overviewed.
Collapse
Affiliation(s)
- Xiaoyang Huang
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zhenhua Zeng
- Biomedical Research Center, Hunan University of Medicine, Huaihua 418000, China
| | - Siqi Li
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Yufei Xie
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyong Tong
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
- Correspondence:
| |
Collapse
|
73
|
Cyclophilin D knockout mice do not accumulate succinate during cardiac ischemia. J Mol Cell Cardiol 2022; 173:73-74. [PMID: 36209553 DOI: 10.1016/j.yjmcc.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/07/2022]
|
74
|
Ojo OO, Ogunleke T, Ajeoge J, Olorunsogo OO. Experimental and molecular docking studies of quercetin and vitamin E with diabetes-associated mitochondrial-ATPase as anti-apoptotic therapeutic strategies. J Diabetes Metab Disord 2022; 21:1717-1729. [PMID: 36404854 PMCID: PMC9672242 DOI: 10.1007/s40200-022-01132-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
Abstract
Purpose Researches have shown the relevance of antioxidants in the management of several diseases. In the present study, the effects of quercetin and vitamin E were investigated on the mitochondrial functions in vivo and in silico. Methods Structures of quercetin and vitamin E were docked against mitochondrial Adenosine triphosphatase (mATPase), and cytochrome c cavity. Activity of liver mATPase and mitochondrial permeability transition pore opening were determined by spectrophotometry and activation of cytochrome c was examined by immunohistochemistry. Results The binding energy of vitamin E (-9 Kcal/mol) in mATPase cavity compares well with glibenclamide (-9.4 Kcal/mol), while quercetin had a binding energy of -7.1 Kcal/mol. Similarly, vitamin E, quercetin were bound to cytochrome c by -6.4 and - 5.5 Kcal/mol energy, while glibenclamide had -7.0 Kcal/mol binding energy. The results showed that vitamin E was more accessible to the protoporphyrin prosthetic group in cytochrome c than quercetin. In the experimental studies, it was validated that vitamin E inhibited the uncontrolled activity of mATPase in diabetic rat liver. This was also proven and tested on the liver mitochondrial permeability transition pore opening observed in diabetic rats. Further experimental assessment of these on activation of cytochrome c showed that vitamin E reduced the extent of the activation more than quercetin and glibenclamide. Conclusion There is a favorable protein-ligand interaction between quercetin and vitamin E in certain apoptotic proteins implicated in diabetes complications.
Collapse
Affiliation(s)
- Oluwatoyin O. Ojo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, 200284 Nigeria
- Department of Chemical Sciences, Faculty of Natural and Applied Sciences, Anchor University Lagos, Lagos, 100278 Nigeria
| | - Titilayo Ogunleke
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, 200284 Nigeria
| | - Joshua Ajeoge
- Department of Chemical Sciences, Faculty of Natural and Applied Sciences, Anchor University Lagos, Lagos, 100278 Nigeria
| | - Olufunso O. Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, 200284 Nigeria
| |
Collapse
|
75
|
Haynes CM, Hekimi S. Mitochondrial dysfunction, aging, and the mitochondrial unfolded protein response in Caenorhabditis elegans. Genetics 2022; 222:iyac160. [PMID: 36342845 PMCID: PMC9713405 DOI: 10.1093/genetics/iyac160] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
We review the findings that establish that perturbations of various aspects of mitochondrial function, including oxidative phosphorylation, can promote lifespan extension, with different types of perturbations acting sometimes independently and additively on extending lifespan. We also review the great variety of processes and mechanisms that together form the mitochondrial unfolded protein response. We then explore the relationships between different types of mitochondrial dysfunction-dependent lifespan extension and the mitochondrial unfolded protein response. We conclude that, although several ways that induce extended lifespan through mitochondrial dysfunction require a functional mitochondrial unfolded protein response, there is no clear indication that activation of the mitochondrial unfolded protein response is sufficient to extend lifespan, despite the fact that the mitochondrial unfolded protein response impacts almost every aspect of mitochondrial function. In fact, in some contexts, mitochondrial unfolded protein response activation is deleterious. To explain this pattern, we hypothesize that, although triggered by mitochondrial dysfunction, the lifespan extension observed might not be the result of a change in mitochondrial function.
Collapse
Affiliation(s)
- Cole M Haynes
- Molecular, Cell and Cancer Biology, UMass-Chan Medical School, Worcester, MA 01655, USA
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
76
|
Sun Y, Zhang P, Li Y, Hou Y, Yin C, Wang Z, Liao Z, Fu X, Li M, Fan C, Sun D, Cheng L. Light-Activated Gold-Selenium Core-Shell Nanocomposites with NIR-II Photoacoustic Imaging Performances for Heart-Targeted Repair. ACS NANO 2022; 16:18667-18681. [PMID: 36264835 DOI: 10.1021/acsnano.2c07311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Mitochondrial dysfunction and oxidative damage represent important pathological mechanisms of myocardial ischemia-reperfusion injury (MI/RI). Searching for potential antioxidant agents to attenuate MI/RI is of great significance in clinic. Herein, gold-selenium core-shell nanostructures (AS-I/S NCs) with good near-infrared (NIR)-II photoacoustic imaging were designed for MI/RI treatment. The AS-I/S NCs after ischemic myocardium-targeted peptide (IMTP) and mitochondrial-targeted antioxidant peptide SS31 modification achieved cardiomyocytes-targeted cellular uptake and enhanced antioxidant ability and significantly inhibited oxygen-glucose deprivation-recovery (OGD/R)-induced cardiotoxicity of H9c2 cells by inhibiting the depletion of mitochondrial membrane potential (MMP) and restoring ATP synthase activity. Furthermore, the AS-I/S NCs after SS31 modification achieved mitochondria-targeted inhibition of reactive oxygen species (ROS) and subsequently attenuated oxidative damage in OGD/R-treated H9c2 cells by inhibition of apoptosis and oxidative damage, regulation of MAPKs and PI3K/AKT pathways. The in vivo AS-I/S NCs administration dramatically improved myocardial functions and angiogenesis and inhibited myocardial fibrosis through inhibiting myocardial apoptosis and oxidative damage in MI/RI of rats. Importantly, the AS-I/S NCs showed good safety and biocompatibility in vivo. Therefore, our findings validated the rational design that mitochondria-targeted selenium-gold nanocomposites could attenuate MI/RI of rats by inhibiting ROS-mediated oxidative damage and regulating MAPKs and PI3K/AKT pathways, which could be a potential therapy for the MI/RI treatment.
Collapse
Affiliation(s)
- Yu Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Pu Zhang
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong 271000, China
| | - Yuqing Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yajun Hou
- Department of Neurology, Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000 Shandong China
| | - Chenyang Yin
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zekun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ziyu Liao
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaoyan Fu
- Department of Neurology, Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000 Shandong China
| | - Man Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Cundong Fan
- Department of Neurology, Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000 Shandong China
| | - Dongdong Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
77
|
Acrolein evokes inflammation and autophagy-dependent apoptosis through oxidative stress in vascular endothelial cells and its protection by 6-C-(E-2-fluorostyryl)naringenin. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
78
|
Wu YJ, Wang SB, Wang LS. SGLT2 Inhibitors: New Hope for the Treatment of Acute Myocardial Infarction? Am J Cardiovasc Drugs 2022; 22:601-613. [PMID: 35947249 DOI: 10.1007/s40256-022-00545-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Among all of the new antidiabetic drugs, an increasing number of studies have evaluated the relationship between the sodium-glucose cotransporter 2 inhibitors (SGLT2i) and acute myocardial infarction (AMI). Since SGLT2i like empagliflozin, canagliflozin, and recently, dapagliflozin have shown impressive positive effects in patients with chronic heart failure with reduced ejection fraction (HFrEF), it has increased research interest to explore the cardiac molecular mechanisms underlying the clinical benefits and attracted more attention to the effects of SGLT2i on a series of cardiovascular events. Experimental and clinical data on SGLT2i treatment after AMI is limited. This is a review of the clinical and preclinical effects of SGLT2i, focusing on available data on the effects of SGLT2i in AMI patients with a brief overview of ongoing trials.
Collapse
Affiliation(s)
- Yu-Jie Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Si-Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Lian-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
79
|
Du M, Li Y, Zhang Q, Zhang J, Ouyang S, Chen Z. The impact of low intensity ultrasound on cells: Underlying mechanisms and current status. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 174:41-49. [PMID: 35764177 DOI: 10.1016/j.pbiomolbio.2022.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 06/10/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Low intensity ultrasound (LIUS) has been adopted for a variety of therapeutic purposes because of its bioeffects such as thermal, mechanical, and cavitation effects. The mechanism of impact and cellular responses of LIUS in cellular regulations have been revealed, which helps to understand the role of LIUS in tumor treatment, stem cell therapy, and nervous system regulation. The review summarizes the bioeffects of LIUS at the cellular level and its related mechanisms, detailing the corresponding theoretical basis and latest research in the study of LIUS in the regulation of cells. In the future, the design of specific LIUS-mediated treatment strategies may benefit from promising investigations which is hoped to provide encouraging therapeutic data.
Collapse
Affiliation(s)
- Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Li
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China; Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qing Zhang
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China; The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Jiaming Zhang
- The First Affiliated Hospital, Center for Reproductive Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuming Ouyang
- The First Affiliated Hospital, Center for Reproductive Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China; The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China.
| |
Collapse
|
80
|
Andelova N, Waczulikova I, Kunstek L, Talian I, Ravingerova T, Jasova M, Suty S, Ferko M. Dichloroacetate as a metabolic modulator of heart mitochondrial proteome under conditions of reduced oxygen utilization. Sci Rep 2022; 12:16348. [PMID: 36175475 PMCID: PMC9522880 DOI: 10.1038/s41598-022-20696-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022] Open
Abstract
Myocardial compensatory mechanisms stimulated by reduced oxygen utilization caused by streptozotocin-induced diabetes mellitus (DM) and treated with dichloroacetate (DCA) are presumably associated with the regulation of mitochondria. We aimed to promote the understanding of key signaling pathways and identify effectors involved in signal transduction. Proteomic analysis and fluorescence spectroscopy measurements revealed significantly decreased membrane potential and upregulated protein amine oxidase [flavin-containing] A (AOFA) in DM mitochondria, indicative of oxidative damage. DCA in diabetic animals (DM + DCA) downregulated AOFA, increased membrane potential, and stimulated thioredoxin-dependent peroxide reductase, a protein with antioxidant function. Furthermore, the DM condition was associated with mitochondrial resistance to calcium overload through mitochondrial permeability transition pores (mPTPs) regulation, despite an increased protein level of voltage-dependent anion-selective protein (VDAC1). In contrast, DM + DCA influenced ROS levels and downregulated VDAC1 and VDAC3 when compared to DM alone. The diabetic myocardium showed an identical pattern of mPTP protein interactions as in the control group, but the interactions were attenuated. Characterization of the combined effect of DM + DCA is a novel finding showing that DCA acted as an effector of VDAC protein interactions, calcium uptake regulation, and ROS production. Overall, DM and DCA did not exhibit an additive effect, but an individual cardioprotective pathway.
Collapse
Affiliation(s)
- Natalia Andelova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Iveta Waczulikova
- Division of Biomedical Physics, Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 84248, Bratislava, Slovakia
| | - Lukas Kunstek
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Ivan Talian
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, 04011, Kosice, Slovakia
| | - Tanya Ravingerova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Magdalena Jasova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Simon Suty
- Division of Biomedical Physics, Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 84248, Bratislava, Slovakia
| | - Miroslav Ferko
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia.
| |
Collapse
|
81
|
Zilio E, Piano V, Wirth B. Mitochondrial Dysfunction in Spinal Muscular Atrophy. Int J Mol Sci 2022; 23:10878. [PMID: 36142791 PMCID: PMC9503857 DOI: 10.3390/ijms231810878] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder caused by recessive mutations in the SMN1 gene, globally affecting ~8-14 newborns per 100,000. The severity of the disease depends on the residual levels of functional survival of motor neuron protein, SMN. SMN is a ubiquitously expressed RNA binding protein involved in a plethora of cellular processes. In this review, we discuss the effects of SMN loss on mitochondrial functions in the neuronal and muscular systems that are the most affected in patients with spinal muscular atrophy. Our aim is to highlight how mitochondrial defects may contribute to disease progression and how restoring mitochondrial functionality may be a promising approach to develop new therapies. We also collected from previous studies a list of transcripts encoding mitochondrial proteins affected in various SMA models. Moreover, we speculate that in adulthood, when motor neurons require only very low SMN levels, the natural deterioration of mitochondria associated with aging may be a crucial triggering factor for adult spinal muscular atrophy, and this requires particular attention for therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Zilio
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Valentina Piano
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
82
|
Alva R, Mirza M, Baiton A, Lazuran L, Samokysh L, Bobinski A, Cowan C, Jaimon A, Obioru D, Al Makhoul T, Stuart JA. Oxygen toxicity: cellular mechanisms in normobaric hyperoxia. Cell Biol Toxicol 2022; 39:111-143. [PMID: 36112262 PMCID: PMC9483325 DOI: 10.1007/s10565-022-09773-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/07/2022] [Indexed: 12/15/2022]
Abstract
In clinical settings, oxygen therapy is administered to preterm neonates and to adults with acute and chronic conditions such as COVID-19, pulmonary fibrosis, sepsis, cardiac arrest, carbon monoxide poisoning, and acute heart failure. In non-clinical settings, divers and astronauts may also receive supplemental oxygen. In addition, under current standard cell culture practices, cells are maintained in atmospheric oxygen, which is several times higher than what most cells experience in vivo. In all the above scenarios, the elevated oxygen levels (hyperoxia) can lead to increased production of reactive oxygen species from mitochondria, NADPH oxidases, and other sources. This can cause cell dysfunction or death. Acute hyperoxia injury impairs various cellular functions, manifesting ultimately as physiological deficits. Chronic hyperoxia, particularly in the neonate, can disrupt development, leading to permanent deficiencies. In this review, we discuss the cellular activities and pathways affected by hyperoxia, as well as strategies that have been developed to ameliorate injury.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Maha Mirza
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Adam Baiton
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lucas Lazuran
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lyuda Samokysh
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ava Bobinski
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Cale Cowan
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Alvin Jaimon
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Dede Obioru
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Tala Al Makhoul
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
83
|
Carew NT, Schmidt HM, Yuan S, Galley JC, Hall R, Altmann HM, Hahn SA, Miller MP, Wood KC, Gabris B, Stapleton MC, Hartwick S, Fazzari M, Wu YL, Trebak M, Kaufman BA, McTiernan CF, Schopfer FJ, Navas P, Thibodeau PH, McNamara DM, Salama G, Straub AC. Loss of cardiomyocyte CYB5R3 impairs redox equilibrium and causes sudden cardiac death. J Clin Invest 2022; 132:e147120. [PMID: 36106636 PMCID: PMC9479700 DOI: 10.1172/jci147120] [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: 01/04/2021] [Accepted: 07/19/2022] [Indexed: 01/04/2023] Open
Abstract
Sudden cardiac death (SCD) in patients with heart failure (HF) is allied with an imbalance in reduction and oxidation (redox) signaling in cardiomyocytes; however, the basic pathways and mechanisms governing redox homeostasis in cardiomyocytes are not fully understood. Here, we show that cytochrome b5 reductase 3 (CYB5R3), an enzyme known to regulate redox signaling in erythrocytes and vascular cells, is essential for cardiomyocyte function. Using a conditional cardiomyocyte-specific CYB5R3-knockout mouse, we discovered that deletion of CYB5R3 in male, but not female, adult cardiomyocytes causes cardiac hypertrophy, bradycardia, and SCD. The increase in SCD in CYB5R3-KO mice is associated with calcium mishandling, ventricular fibrillation, and cardiomyocyte hypertrophy. Molecular studies reveal that CYB5R3-KO hearts display decreased adenosine triphosphate (ATP), increased oxidative stress, suppressed coenzyme Q levels, and hemoprotein dysregulation. Finally, from a translational perspective, we reveal that the high-frequency missense genetic variant rs1800457, which translates into a CYB5R3 T117S partial loss-of-function protein, associates with decreased event-free survival (~20%) in Black persons with HF with reduced ejection fraction (HFrEF). Together, these studies reveal a crucial role for CYB5R3 in cardiomyocyte redox biology and identify a genetic biomarker for persons of African ancestry that may potentially increase the risk of death from HFrEF.
Collapse
Affiliation(s)
- Nolan T. Carew
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Heidi M. Schmidt
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute
| | - Joseph C. Galley
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Robert Hall
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | | | | | | | - Katherine C. Wood
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Bethann Gabris
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margaret C. Stapleton
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sean Hartwick
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Yijen L. Wu
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Brett A. Kaufman
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles F. McTiernan
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco J. Schopfer
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Placido Navas
- Andalusian Center for Developmental Biology and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | | | - Dennis M. McNamara
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Guy Salama
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
- Center for Microvascular Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
84
|
Carbone F, Liberale L, Preda A, Schindler TH, Montecucco F. Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting. Cells 2022; 11:2833. [PMID: 36139408 PMCID: PMC9496713 DOI: 10.3390/cells11182833] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
The onset of cardiomyopathy is a common feature in sepsis, with relevant effects on its pathophysiology and clinical care. Septic cardiomyopathy is characterized by reduced left ventricular (LV) contractility eventually associated with LV dilatation with or without right ventricle failure. Unfortunately, such a wide range of ultrasonographic findings does not reflect a deep comprehension of sepsis-induced cardiomyopathy, but rather a lack of consensus about its definition. Several echocardiographic parameters intrinsically depend on loading conditions (both preload and afterload) so that it may be challenging to discriminate which is primitive and which is induced by hemodynamic perturbances. Here, we explore the state of the art in sepsis-related cardiomyopathy. We focus on the shortcomings in its definition and point out how cardiac performance dynamically changes in response to different hemodynamic clusters. A special attention is also given to update the knowledge about molecular mechanisms leading to myocardial dysfunction and that recall those of myocardial hibernation. Ultimately, the aim of this review is to highlight the unsolved issue in the field of sepsis-induced cardiomyopathy as their implementation would lead to improve risk stratification and clinical care.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Alberto Preda
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Thomas Hellmut Schindler
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| |
Collapse
|
85
|
Affiliation(s)
- Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.)
| |
Collapse
|
86
|
Sandroni PB, Fisher-Wellman KH, Jensen BC. Adrenergic Receptor Regulation of Mitochondrial Function in Cardiomyocytes. J Cardiovasc Pharmacol 2022; 80:364-377. [PMID: 35170492 PMCID: PMC9365878 DOI: 10.1097/fjc.0000000000001241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Adrenergic receptors (ARs) are G protein-coupled receptors that are stimulated by catecholamines to induce a wide array of physiological effects across tissue types. Both α1- and β-ARs are found on cardiomyocytes and regulate cardiac contractility and hypertrophy through diverse molecular pathways. Acute activation of cardiomyocyte β-ARs increases heart rate and contractility as an adaptive stress response. However, chronic β-AR stimulation contributes to the pathobiology of heart failure. By contrast, mounting evidence suggests that α1-ARs serve protective functions that may mitigate the deleterious effects of chronic β-AR activation. Here, we will review recent studies demonstrating that α1- and β-ARs differentially regulate mitochondrial biogenesis and dynamics, mitochondrial calcium handling, and oxidative phosphorylation in cardiomyocytes. We will identify potential mechanisms of these actions and focus on the implications of these findings for the modulation of contractile function in the uninjured and failing heart. Collectively, we hope to elucidate important physiological processes through which these well-studied and clinically relevant receptors stimulate and fuel cardiac contraction to contribute to myocardial health and disease.
Collapse
Affiliation(s)
- Peyton B. Sandroni
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
| | - Kelsey H. Fisher-Wellman
- East Carolina University Brody School of Medicine, Department of Physiology
- East Carolina University Diabetes and Obesity Institute
| | - Brian C. Jensen
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
- University of North Carolina School of Medicine, Department of Medicine, Division of Cardiology
| |
Collapse
|
87
|
Li Z, Zhang J, Duan X, Zhao G, Zhang M. Celastrol: A Promising Agent Fighting against Cardiovascular Diseases. Antioxidants (Basel) 2022; 11:antiox11081597. [PMID: 36009315 PMCID: PMC9405053 DOI: 10.3390/antiox11081597] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases (CVD) are leading causes of morbidity and mortality worldwide; therefore, seeking effective therapeutics to reduce the global burden of CVD has become increasingly urgent. Celastrol, a bioactive compound isolated from the roots of the plant Tripterygium wilfordii (TW), has been attracting increasing research attention in recent years, as it exerts cardiovascular treatment benefits targeting both CVD and their associated risk factors. Substantial evidence has revealed a protective role of celastrol against a broad spectrum of CVD including obesity, diabetes, atherosclerosis, cerebrovascular injury, calcific aortic valve disease and heart failure through complicated and interlinked mechanisms such as direct protection against cardiomyocyte hypertrophy and death, and indirect action on oxidation and inflammation. This review will mainly summarize the beneficial effects of celastrol against CVD, largely based on in vitro and in vivo preclinical studies, and the potential underlying mechanisms. We will also briefly discuss celastrol’s pharmacokinetic limitations, which hamper its further clinical applications, and prospective future directions.
Collapse
Affiliation(s)
- Zhexi Li
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Jingyi Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Research Excellence, London SE5 9NU, UK
| | - Xulei Duan
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Guoan Zhao
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Min Zhang
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Research Excellence, London SE5 9NU, UK
- Correspondence: ; Tel.: +44-207848-5319; Fax: +44-207848-5193
| |
Collapse
|
88
|
Stamerra CA, Di Giosia P, Giorgini P, Ferri C, Sukhorukov VN, Sahebkar A. Mitochondrial Dysfunction and Cardiovascular Disease: Pathophysiology and Emerging Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9530007. [PMID: 35958017 PMCID: PMC9363184 DOI: 10.1155/2022/9530007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Mitochondria ensure the supply of cellular energy through the production of ATP via oxidative phosphorylation. The alteration of this process, called mitochondrial dysfunction, leads to a reduction in ATP and an increase in the production of reactive oxygen species (ROS). Mitochondrial dysfunction can be caused by mitochondrial/nuclear DNA mutations, or it can be secondary to pathological conditions such as cardiovascular disease, aging, and environmental stress. The use of therapies aimed at the prevention/correction of mitochondrial dysfunction, in the context of the specific treatment of cardiovascular diseases, is a topic of growing interest. In this context, the data are conflicting since preclinical studies are numerous, but there are no large randomized studies.
Collapse
Affiliation(s)
- Cosimo Andrea Stamerra
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Di Giosia
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Giorgini
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Claudio Ferri
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
89
|
Milliken AS, Nadtochiy SM, Brookes PS. Inhibiting Succinate Release Worsens Cardiac Reperfusion Injury by Enhancing Mitochondrial Reactive Oxygen Species Generation. J Am Heart Assoc 2022; 11:e026135. [PMID: 35766275 PMCID: PMC9333399 DOI: 10.1161/jaha.122.026135] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background The metabolite succinate accumulates during cardiac ischemia. Within 5 minutes of reperfusion, succinate returns to baseline levels via both its release from cells and oxidation by mitochondrial complex II. The latter drives reactive oxygen species (ROS) generation and subsequent opening of the mitochondrial permeability transition (PT) pore, leading to cell death. Targeting succinate dynamics (accumulation/oxidation/release) may be therapeutically beneficial in cardiac ischemia–reperfusion (IR) injury. It has been proposed that blocking MCT1 (monocarboxylate transporter 1) may be beneficial in IR injury, by preventing succinate release and subsequent engagement of downstream inflammatory signaling pathways. In contrast, herein we hypothesized that blocking MCT1 would retain succinate in cells, exacerbating ROS generation and IR injury. Methods and Results Using the mitochondrial ROS probe mitoSOX and a custom‐built murine heart perfusion rig built into a spectrofluorometer, we measured ROS generation in situ during the first moments of reperfusion. We found that acute MCT1 inhibition enhanced mitochondrial ROS generation at reperfusion and worsened IR injury (recovery of function and infarct size). Both of these effects were abrogated by tandem inhibition of mitochondrial complex II, suggesting that succinate retention worsens IR because it drives more mitochondrial ROS generation. Furthermore, using the PT pore inhibitor cyclosporin A, along with monitoring of PT pore opening via the mitochondrial membrane potential indicator tetramethylrhodamine ethyl ester, we herein provide evidence that ROS generation during early reperfusion is upstream of the PT pore, not downstream as proposed by others. In addition, pore opening was exacerbated by MCT1 inhibition. Conclusions Together, these findings highlight the importance of succinate dynamics and mitochondrial ROS generation as key determinants of PT pore opening and IR injury outcomes.
Collapse
Affiliation(s)
- Alexander S Milliken
- Department of Pharmacology and Physiology University of Rochester Medical Center Rochester NY
| | - Sergiy M Nadtochiy
- Department of Anesthesiology and Perioperative Medicine University of Rochester Medical Center Rochester NY
| | - Paul S Brookes
- Department of Anesthesiology and Perioperative Medicine University of Rochester Medical Center Rochester NY
| |
Collapse
|
90
|
Tricaud N, Gautier B, Berthelot J, Gonzalez S, Van Hameren G. Traumatic and Diabetic Schwann Cell Demyelination Is Triggered by a Transient Mitochondrial Calcium Release through Voltage Dependent Anion Channel 1. Biomedicines 2022; 10:biomedicines10061447. [PMID: 35740468 PMCID: PMC9220872 DOI: 10.3390/biomedicines10061447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
A large number of peripheral neuropathies, among which are traumatic and diabetic peripheral neuropathies, result from the degeneration of the myelin sheath, a process called demyelination. Demyelination does not result from Schwann cell death but from Schwann cell dedifferentiation, which includes reprograming and several catabolic and anabolic events. Starting around 4 h after nerve injury, activation of MAPK/cJun pathways is the earliest characterized step of this dedifferentiation program. Here we show, using real-time in vivo imaging, that Schwann cell mitochondrial pH, motility and calcium content are altered as soon as one hour after nerve injury. Mitochondrial calcium release occurred through the VDAC outer membrane channel and mPTP inner membrane channel. This calcium influx in the cytoplasm induced Schwann-cell demyelination via MAPK/c-Jun activation. Blocking calcium release through VDAC silencing or VDAC inhibitor TRO19622 prevented demyelination. We found that the kinetics of mitochondrial calcium release upon nerve injury were altered in the Schwann cells of diabetic mice suggesting a permanent leak of mitochondrial calcium in the cytoplasm. TRO19622 treatment alleviated peripheral nerve defects and motor deficit in diabetic mice. Together, these data indicate that mitochondrial calcium homeostasis is instrumental in the Schwann cell demyelination program and that blocking VDAC constitutes a molecular basis for developing anti-demyelinating drugs for diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Nicolas Tricaud
- Institut des Neurosciences de Montpellier, Univ. Montpellier, INSERM, 34000 Montpellier, France; (B.G.); (J.B.); (S.G.); (G.V.H.)
- I-Stem, UEVE/UPS U861, INSERM U861, AFM, 91100 Corbeil-Essonnes, France
- Correspondence:
| | - Benoit Gautier
- Institut des Neurosciences de Montpellier, Univ. Montpellier, INSERM, 34000 Montpellier, France; (B.G.); (J.B.); (S.G.); (G.V.H.)
| | - Jade Berthelot
- Institut des Neurosciences de Montpellier, Univ. Montpellier, INSERM, 34000 Montpellier, France; (B.G.); (J.B.); (S.G.); (G.V.H.)
| | - Sergio Gonzalez
- Institut des Neurosciences de Montpellier, Univ. Montpellier, INSERM, 34000 Montpellier, France; (B.G.); (J.B.); (S.G.); (G.V.H.)
| | - Gerben Van Hameren
- Institut des Neurosciences de Montpellier, Univ. Montpellier, INSERM, 34000 Montpellier, France; (B.G.); (J.B.); (S.G.); (G.V.H.)
| |
Collapse
|
91
|
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells 2022; 11:cells11111855. [PMID: 35681549 PMCID: PMC9180016 DOI: 10.3390/cells11111855] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
p66Shc is a widely expressed protein that governs a variety of cardiovascular pathologies by generating, and exacerbating, pro-apoptotic ROS signals. Here, we review p66Shc’s connections to reactive oxygen species, expression, localization, and discuss p66Shc signaling and mitochondrial functions. Emphasis is placed on recent p66Shc mitochondrial function discoveries including structure/function relationships, ROS identity and regulation, mechanistic insights, and how p66Shc-cyt c interactions can influence p66Shc mitochondrial function. Based on recent findings, a new p66Shc mitochondrial function model is also put forth wherein p66Shc acts as a rheostat that can promote or antagonize apoptosis. A discussion of how the revised p66Shc model fits previous findings in p66Shc-mediated cardiovascular pathology follows.
Collapse
Affiliation(s)
- Landon Haslem
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Jennifer M. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Franklin A. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
- Stephenson Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
92
|
Chapa-Dubocq XR, Garcia-Baez JF, Bazil JN, Javadov S. Crosstalk between adenine nucleotide transporter and mitochondrial swelling: experimental and computational approaches. Cell Biol Toxicol 2022:10.1007/s10565-022-09724-2. [PMID: 35606662 DOI: 10.1007/s10565-022-09724-2] [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: 01/20/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
Mitochondrial metabolism and function are modulated by changes in matrix Ca2+. Small increases in the matrix Ca2+ stimulate mitochondrial bioenergetics, whereas excessive Ca2+ leads to cell death by causing massive matrix swelling and impairing the structural and functional integrity of mitochondria. Sustained opening of the non-selective mitochondrial permeability transition pores (PTP) is the main mechanism responsible for mitochondrial Ca2+ overload that leads to mitochondrial dysfunction and cell death. Recent studies suggest the existence of two or more types of PTP, and adenine nucleotide translocator (ANT) and FOF1-ATP synthase were proposed to form the PTP independent of each other. Here, we elucidated the role of ANT in PTP opening by applying both experimental and computational approaches. We first developed and corroborated a detailed model of the ANT transport mechanism including the matrix (ANTM), cytosolic (ANTC), and pore (ANTP) states of the transporter. Then, the ANT model was incorporated into a simple, yet effective, empirical model of mitochondrial bioenergetics to ascertain the point when Ca2+ overload initiates PTP opening via an ANT switch-like mechanism activated by matrix Ca2+ and is inhibited by extra-mitochondrial ADP. We found that encoding a heterogeneous Ca2+ response of at least three types of PTPs, weakly, moderately, and strongly sensitive to Ca2+, enabled the model to simulate Ca2+ release dynamics observed after large boluses were administered to a population of energized cardiac mitochondria. Thus, this study demonstrates the potential role of ANT in PTP gating and proposes a novel mechanism governing the cryptic nature of the PTP phenomenon.
Collapse
Affiliation(s)
- Xavier R Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA
| | - Jorge F Garcia-Baez
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA
| | - Jason N Bazil
- Department of Physiology, Michigan State University, East Lansing, MI, 48824-1046, USA
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA.
| |
Collapse
|
93
|
Castilla R, Ruffa FV, Bancalari I, Fernández Vivanco M, Lallopizzo C, Torasso N, Farcy N, Gutierrez C, Bonazzolaa P. Cobalt chloride postconditioning as myoprotective therapy in cardiac ischemia-reperfusion. Pflugers Arch 2022; 474:743-752. [PMID: 35585327 DOI: 10.1007/s00424-022-02703-w] [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: 01/10/2022] [Revised: 03/11/2022] [Accepted: 05/03/2022] [Indexed: 11/24/2022]
Abstract
Since damage induced by ischemia-reperfusion (I/R) involves alterations in Ca2+ homeostasis and is reduced by ischemic postconditioning (IP) and that CoCl2 can trigger changes resembling the response to a hypoxic event in normoxia and its blockade on Ca2+ current in heart muscle, our aim was to evaluate CoCl2 as an IP therapeutic tool. Mechanic and energetic parameters of isolated and arterially perfused male Wistar rat heart ventricles were simultaneously analyzed in a model of I/R in which 0.23 mmol/L CoCl2 was introduced upon reperfusion and kept or withdrawn after 20 min or introduced after 20 min of reperfusion. The presence of CoCl2 did not affect diastolic pressure but increased post-ischemic contractile recovery, which peaked at 20 min and decreased at the end of reperfusion. This decrease was prevented when CoCl2 was removed at 20 min of reperfusion. Total heat release increased throughout reperfusion, while economy increased between 15 and 25 min. No effect was observed when CoCl2 was introduced at 20 min of reperfusion. In addition, both the area under the contracture curve evoked by 10 mmol/L caffeine-36 mmol/L Na+ and the contracture tension relaxation rate were higher with CoCl2.Furthermore, CoCl2 decreased the number of arrhythmias during reperfusion and the ventricular damaged area. The presence of CoCl2 in reperfusion induces cardioprotection consistent with the improvement in cellular calcium handling. The use of CoCl2 constitutes a potential cardioprotective tool of clinical relevance.
Collapse
Affiliation(s)
- Rocío Castilla
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina.
| | - Facundo Vigón Ruffa
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Ignacio Bancalari
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Mercedes Fernández Vivanco
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Carla Lallopizzo
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Nicolás Torasso
- Facultad de Ciencias Exactas Y Naturales, Instituto de Física de Buenos Aires (IFIBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicole Farcy
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Christopher Gutierrez
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| | - Patricia Bonazzolaa
- CONICET, Instituto Alberto C Taquini de Investigaciones en Medicina Traslacional (IATIMET) C1122AAJ, Universidad de Buenos Aires, Marcelo T. de Alvear, 2270- C1122AAJ, Buenos Aires, Argentina
| |
Collapse
|
94
|
Mitochondrial Damage in Myocardial Ischemia/Reperfusion Injury and Application of Natural Plant Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8726564. [PMID: 35615579 PMCID: PMC9126658 DOI: 10.1155/2022/8726564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/28/2022]
Abstract
Ischemic heart disease (IHD) is currently one of the leading causes of death among cardiovascular diseases worldwide. In addition, blood reflow and reperfusion paradoxically also lead to further death of cardiomyocytes and increase the infarct size. Multiple evidences indicated that mitochondrial function and structural disorders were the basic driving force of IHD. We summed up the latest evidence of the basic associations and underlying mechanisms of mitochondrial damage in the event of ischemia/reperfusion (I/R) injury. This review then reviewed natural plant products (NPPs) which have been demonstrated to mitochondria-targeted therapeutic effects during I/R injury and the potential pathways involved. We realized that NPPs mainly maintained the integrality of mitochondria membrane and ameliorated dysfunction, such as improving abnormal mitochondrial calcium handling and inhibiting oxidative stress, so as to protect cardiomyocytes during I/R injury. This information will improve our knowledge of mitochondrial biology and I/R-induced injury's pathogenesis and exhibit that NPPs hold promise for translation into potential therapies that target mitochondria.
Collapse
|
95
|
Li X, Flynn ER, do Carmo JM, Wang Z, da Silva AA, Mouton AJ, Omoto ACM, Hall ME, Hall JE. Direct Cardiac Actions of Sodium-Glucose Cotransporter 2 Inhibition Improve Mitochondrial Function and Attenuate Oxidative Stress in Pressure Overload-Induced Heart Failure. Front Cardiovasc Med 2022; 9:859253. [PMID: 35647080 PMCID: PMC9135142 DOI: 10.3389/fcvm.2022.859253] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/15/2022] [Indexed: 12/21/2022] Open
Abstract
Clinical trials showed that sodium-glucose cotransporter 2 (SGLT2) inhibitors, a class of drugs developed for treating diabetes mellitus, improve prognosis of patients with heart failure (HF). However, the mechanisms for cardioprotection by SGLT2 inhibitors are still unclear. Mitochondrial dysfunction and oxidative stress play important roles in progression of HF. This study tested the hypothesis that empagliflozin (EMPA), a highly selective SGLT2 inhibitor, improves mitochondrial function and reduces reactive oxygen species (ROS) while enhancing cardiac performance through direct effects on the heart in a non-diabetic mouse model of HF induced by transverse aortic constriction (TAC). EMPA or vehicle was administered orally for 4 weeks starting 2 weeks post-TAC. EMPA treatment did not alter blood glucose or body weight but significantly attenuated TAC-induced cardiac dysfunction and ventricular remodeling. Impaired mitochondrial oxidative phosphorylation (OXPHOS) in failing hearts was significantly improved by EMPA. EMPA treatment also enhanced mitochondrial biogenesis and restored normal mitochondria morphology. Although TAC increased mitochondrial ROS and decreased endogenous antioxidants, EMPA markedly inhibited cardiac ROS production and upregulated expression of endogenous antioxidants. In addition, EMPA enhanced autophagy and decreased cardiac apoptosis in TAC-induced HF. Importantly, mitochondrial respiration significantly increased in ex vivo cardiac fibers after direct treatment with EMPA. Our results indicate that EMPA has direct effects on the heart, independently of reductions in blood glucose, to enhance mitochondrial function by upregulating mitochondrial biogenesis, enhancing OXPHOS, reducing ROS production, attenuating apoptosis, and increasing autophagy to improve overall cardiac function in a non-diabetic model of pressure overload-induced HF.
Collapse
Affiliation(s)
- Xuan Li
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Serrat R, Oliveira-Pinto A, Marsicano G, Pouvreau S. Imaging mitochondrial calcium dynamics in the central nervous system. J Neurosci Methods 2022; 373:109560. [PMID: 35320763 DOI: 10.1016/j.jneumeth.2022.109560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 12/28/2022]
Abstract
Mitochondrial calcium handling is a particularly active research area in the neuroscience field, as it plays key roles in the regulation of several functions of the central nervous system, such as synaptic transmission and plasticity, astrocyte calcium signaling, neuronal activity… In the last few decades, a panel of techniques have been developed to measure mitochondrial calcium dynamics, relying mostly on photonic microscopy, and including synthetic sensors, hybrid sensors and genetically encoded calcium sensors. The goal of this review is to endow the reader with a deep knowledge of the historical and latest tools to monitor mitochondrial calcium events in the brain, as well as a comprehensive overview of the current state of the art in brain mitochondrial calcium signaling. We will discuss the main calcium probes used in the field, their mitochondrial targeting strategies, their key properties and major drawbacks. In addition, we will detail the main roles of mitochondrial calcium handling in neuronal tissues through an extended report of the recent studies using mitochondrial targeted calcium sensors in neuronal and astroglial cells, in vitro and in vivo.
Collapse
Affiliation(s)
- Roman Serrat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Alexandre Oliveira-Pinto
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Giovanni Marsicano
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Sandrine Pouvreau
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
97
|
Preserved Left Ventricular Function despite Myocardial Fibrosis and Myopathy in the Dystrophin-Deficient D2.B10-Dmdmdx/J Mouse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5362115. [PMID: 35340200 PMCID: PMC8942668 DOI: 10.1155/2022/5362115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 11/18/2022]
Abstract
Duchenne muscular dystrophy involves an absence of dystrophin, a cytoskeletal protein which supports cell structural integrity and scaffolding for signalling molecules in myocytes. Affected individuals experience progressive muscle degeneration that leads to irreversible loss of ambulation and respiratory diaphragm function. Although clinical management has greatly advanced, heart failure due to myocardial cell loss and fibrosis remains the major cause of death. We examined cardiac morphology and function in D2.B10-Dmdmdx/J (D2-mdx) mice, a relatively new mouse model of muscular dystrophy, which we compared to their wild-type background DBA/2J mice (DBA/2). We also tested whether drug treatment with a specific blocker of mitochondrial permeability transition pore opening (Debio-025), or ACE inhibition (Perindopril), had any effect on dystrophy-related cardiomyopathy. D2-mdx mice were treated for six weeks with Vehicle control, Debio-025 (20 mg/kg/day), Perindopril (2 mg/kg/day), or a combination (n = 8/group). At 18 weeks, compared to DBA/2, D2-mdx hearts displayed greater ventricular collagen, lower cell density, greater cell diameter, and greater protein expression levels of IL-6, TLR4, BAX/Bcl2, caspase-3, PGC-1α, and notably monoamine oxidases A and B. Remarkably, these adaptations in D2-mdx mice were associated with preserved resting left ventricular function similar to DBA/2 mice. Compared to vehicle, although Perindopril partly attenuated the increase in heart weight and collagen at 18 weeks, the drug treatments had no marked impact on dystrophic cardiomyopathy.
Collapse
|
98
|
Abstract
T lymphocytes (T cells) are divided into two functionally different subgroups the CD4+ T helper cells (Th) and the CD8+ cytotoxic T lymphocytes (CTL). Adequate CD4 and CD8 T cell activation to proliferation, clonal expansion and effector function is crucial for efficient clearance of infection by pathogens. Failure to do so may lead to T cell exhaustion. Upon activation by antigen presenting cells, T cells undergo metabolic reprograming that support effector functions. In this review we will discuss how metabolic reprograming dictates functionality during viral infections using severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human immunodeficiency virus (HIV) as examples. Moreover, we will briefly discuss T cell metabolic programs during bacterial infections exemplified by Mycobacterium tuberculosis (MT) infection.
Collapse
Affiliation(s)
| | - Bjørn Steen Skålhegg
- Division for Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
99
|
Villavicencio-Tejo F, Olesen MA, Aránguiz A, Quintanilla RA. Activation of the Nrf2 Pathway Prevents Mitochondrial Dysfunction Induced by Caspase-3 Cleaved Tau: Implications for Alzheimer’s Disease. Antioxidants (Basel) 2022; 11:antiox11030515. [PMID: 35326165 PMCID: PMC8944569 DOI: 10.3390/antiox11030515] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by memory and cognitive impairment, accompanied by the accumulation of extracellular deposits of amyloid β-peptide (Aβ) and the presence of neurofibrillary tangles (NFTs) composed of pathological forms of tau protein. Mitochondrial dysfunction and oxidative stress are also critical elements for AD development. We previously showed that the presence of caspase-3 cleaved tau, a relevant pathological form of tau in AD, induced mitochondrial dysfunction and oxidative damage in different neuronal models. Recent studies demonstrated that the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) plays a significant role in the antioxidant response promoting neuroprotection. Here, we studied the effects of Nrf2 activation using sulforaphane (SFN) against mitochondrial injury induced by caspase-3 cleaved tau. We used immortalized cortical neurons to evaluate mitochondrial bioenergetics and ROS levels in control and SFN-treated cells. Expression of caspase-3 cleaved tau induced mitochondrial fragmentation, depolarization, ATP loss, and increased ROS levels. Treatment with SFN for 24 h significantly prevented these mitochondrial abnormalities, and reduced ROS levels. Analysis of Western blots and rt-PCR studies showed that SFN treatment increased the expression of several Nrf2-related antioxidants genes in caspase-3 cleaved tau cells. These results indicate a potential role of the Nrf2 pathway in preventing mitochondrial dysfunction induced by pathological forms of tau in AD.
Collapse
|
100
|
Vlasov AV, Osipov SD, Bondarev NA, Uversky VN, Borshchevskiy VI, Yanyushin MF, Manukhov IV, Rogachev AV, Vlasova AD, Ilyinsky NS, Kuklin AI, Dencher NA, Gordeliy VI. ATP synthase F OF 1 structure, function, and structure-based drug design. Cell Mol Life Sci 2022; 79:179. [PMID: 35253091 PMCID: PMC11072866 DOI: 10.1007/s00018-022-04153-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/26/2021] [Accepted: 01/14/2022] [Indexed: 11/30/2022]
Abstract
ATP synthases are unique rotatory molecular machines that supply biochemical reactions with adenosine triphosphate (ATP)-the universal "currency", which cells use for synthesis of vital molecules and sustaining life. ATP synthases of F-type (FOF1) are found embedded in bacterial cellular membrane, in thylakoid membranes of chloroplasts, and in mitochondrial inner membranes in eukaryotes. The main functions of ATP synthases are control of the ATP synthesis and transmembrane potential. Although the key subunits of the enzyme remain highly conserved, subunit composition and structural organization of ATP synthases and their assemblies are significantly different. In addition, there are hypotheses that the enzyme might be involved in the formation of the mitochondrial permeability transition pore and play a role in regulation of the cell death processes. Dysfunctions of this enzyme lead to numerous severe disorders with high fatality levels. In our review, we focus on FOF1-structure-based approach towards development of new therapies by using FOF1 structural features inherited by the representatives of this enzyme family from different taxonomy groups. We analyzed and systematized the most relevant information about the structural organization of FOF1 to discuss how this approach might help in the development of new therapies targeting ATP synthases and design tools for cellular bioenergetics control.
Collapse
Affiliation(s)
- Alexey V Vlasov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Stepan D Osipov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Nikolay A Bondarev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Vladimir N Uversky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Valentin I Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425, Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428, Jülich, Germany
| | - Mikhail F Yanyushin
- Institute of Basic Biological Problems, Russian Academy of Sciences, 142290, Pushchino, Moscow region, Russia
| | - Ilya V Manukhov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Andrey V Rogachev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Anastasiia D Vlasova
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Nikolay S Ilyinsky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Alexandr I Kuklin
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Norbert A Dencher
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Physical Biochemistry, Department Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 4, 64287, Darmstadt, Germany
| | - Valentin I Gordeliy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia.
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425, Jülich, Germany.
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428, Jülich, Germany.
- Institut de Biologie Structurale Jean-Pierre Ebel, Université Grenoble Alpes-Commissariat à l'Energie Atomique et aux Energies Alternatives-CNRS, 38027, Grenoble, France.
| |
Collapse
|