1
|
Mattiazzi A, Jaquenod De Giusti C, Valverde CA. CaMKII at the crossroads: calcium dysregulation, and post-translational modifications driving cell death. J Physiol 2025. [PMID: 39907446 DOI: 10.1113/jp285941] [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: 10/16/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025] Open
Abstract
The multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates numerous proteins involved in excitation-contraction-relaxation coupling and cardiac excitability. However, its overactivation induces severe Ca2+/handling alterations, playing a significant role in the pathogenesis of diseases such as hypertrophy, arrhythmias and cell death, which can ultimately lead to heart failure. Being a suitable target for various aberrant signals that characterize several diseases, such as Ca2+ overload, oxidative stress or excessive glycosylation, CaMKII shifts under these conditions from a physiological regulator to a pathological molecule. In this review, we explore the evolution of knowledge regarding the role of CaMKII activation on cell death across different pathological contexts, focusing on the converging mechanisms that transform the enzyme from an ally into a villain.
Collapse
Affiliation(s)
- Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares 'Dr Horacio E. Cingolani,' CCT-La Plata/CONICET, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares 'Dr Horacio E. Cingolani,' CCT-La Plata/CONICET, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares 'Dr Horacio E. Cingolani,' CCT-La Plata/CONICET, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| |
Collapse
|
2
|
Lee KCY, Williams AL, Wang L, Xie G, Jia W, Fujimoto A, Gerschenson M, Shohet RV. PKM2 regulates metabolic flux and oxidative stress in the murine heart. Physiol Rep 2024; 12:e70040. [PMID: 39256891 PMCID: PMC11387154 DOI: 10.14814/phy2.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Cardiac metabolism ensures a continuous ATP supply, primarily using fatty acids in a healthy state and favoring glucose in pathological conditions. Pyruvate kinase muscle (PKM) controls the final step of glycolysis, with PKM1 being the main isoform in the heart. PKM2, elevated in various heart diseases, has been suggested to play a protective role in cardiac stress, but its function in basal cardiac metabolism remains unclear. We examined hearts from global PKM2 knockout (PKM2-/-) mice and found reduced intracellular glucose. Isotopic tracing of U-13C glucose revealed a shift to biosynthetic pathways in PKM2-/- cardiomyocytes. Total ATP content was two-thirds lower in PKM2-/- hearts, and functional analysis indicated reduced mitochondrial oxygen consumption. Total reactive oxygen species (ROS) and mitochondrial superoxide were also increased in PKM2-/- cardiomyocytes. Intriguingly, PKM2-/- hearts had preserved ejection fraction compared to controls. Mechanistically, increased calcium/calmodulin-dependent kinase II activity and phospholamban phosphorylation may contribute to higher sarcoendoplasmic reticulum calcium ATPase 2 pump activity in PKM2-/- hearts. Loss of PKM2 led to altered glucose metabolism, diminished mitochondrial function, and increased ROS in cardiomyocytes. These data suggest that cardiac PKM2 acts as an important rheostat to maintain ATP levels while limiting oxidative stress. Although loss of PKM2 did not impair baseline contractility, its absence may make hearts more sensitive to environmental stress or injury.
Collapse
Affiliation(s)
- Katie C. Y. Lee
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Allison L. Williams
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Lu Wang
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Guoxiang Xie
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Wei Jia
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Anastasia Fujimoto
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Ralph V. Shohet
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| |
Collapse
|
3
|
Pullara F, Forsmann MC, General IJ, Ayoob JC, Furbee E, Castro SL, Hu X, Greenamyre JT, Di Maio R. NADPH oxidase 2 activity disrupts Calmodulin/CaMKIIα complex via redox modifications of CaMKIIα-contained Cys30 and Cys289: Implications in Parkinson's disease. Redox Biol 2024; 75:103254. [PMID: 38968922 PMCID: PMC11278932 DOI: 10.1016/j.redox.2024.103254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 07/07/2024] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II α (CaMKIIα) signaling in the brain plays a critical role in regulating neuronal Ca2+ homeostasis. Its dysfunctional activity is associated with various neurological and neurodegenerative disorders, including Parkinson's disease (PD). Using computational modeling analysis, we predicted that, two essential cysteine residues contained in CaMKIIα, Cys30 and Cys289, may undergo redox modifications impacting the proper functioning of the CaMKIIα docking site for Ca2+/CaM, thus impeding the formation of the CaMKIIα:Ca2+/CaM complex, essential for a proper modulation of CaMKIIα kinase activity. Our subsequent in vitro investigations confirmed the computational predictions, specifically implicating Cys30 and Cys289 residues in impairing CaMKIIα:Ca2+/CaM interaction. We observed CaMKIIα:Ca2+/CaM complex disruption in dopamine (DA) nigrostriatal neurons of post-mortem Parkinson's disease (PD) patients' specimens, addressing the high relevance of this event in the disease. CaMKIIα:Ca2+/CaM complex disruption was also observed in both in vitro and in vivo rotenone models of PD, where this phenomenon was associated with CaMKIIα kinase hyperactivity. Moreover, we observed that, NADPH oxidase 2 (NOX2), a major enzymatic generator of superoxide anion (O2●-) and hydrogen peroxide (H2O2) in the brain with implications in PD pathogenesis, is responsible for CaMKIIα:Ca2+/CaM complex disruption associated to a stable Ca2+CAM-independent CaMKIIα kinase activity and intracellular Ca2+ accumulation. The present study highlights the importance of oxidative stress, in disturbing the delicate balance of CaMKIIα signaling in calcium dysregulation, offering novel insights into PD pathogenesis.
Collapse
Affiliation(s)
| | - Madison C Forsmann
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Ignacio J General
- School of Science and Technology, Universidad Nacional de San Martin, San Martín, 1650, Buenos Aires, Argentina
| | - Joseph C Ayoob
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Emily Furbee
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sandra L Castro
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Xiaoping Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - J Timothy Greenamyre
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA
| | - Roberto Di Maio
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
4
|
Gao Y, Li S, Liu X, Si D, Chen W, Yang F, Sun H, Yang P. RyR2 Stabilizer Attenuates Cardiac Hypertrophy by Downregulating TNF-α/NF-κB/NLRP3 Signaling Pathway through Inhibiting Calcineurin. J Cardiovasc Transl Res 2024; 17:481-495. [PMID: 38652413 DOI: 10.1007/s12265-023-10376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/13/2023] [Indexed: 04/25/2024]
Abstract
The effect of Ryanodine receptor2 (RyR2) and its stabilizer on cardiac hypertrophy is not well known. C57/BL6 mice underwent transverse aortic contraction (TAC) or sham surgery were administered dantrolene, the RyR2 stabilizer, or control drug. Dantrolene significantly alleviated TAC-induced cardiac hypertrophy in mice, and RNA sequencing was performed implying calcineurin/NFAT3 and TNF-α/NF-κB/NLRP3 as critical signaling pathways. Further expression analysis and Western blot with heart tissue as well as neonatal rat cardiomyocyte (NRCM) model confirmed dantrolene decreases the activation of calcineurin/NFAT3 signaling pathway and TNF-α/NF-κB/NLRP3 signaling pathway, which was similar to FK506 and might be attenuated by calcineurin overexpression. The present study shows for the first time that RyR2 stabilizer dantrolene attenuates cardiac hypertrophy by inhibiting the calcineurin, therefore downregulating the TNF-α/NF-κB/NLRP3 pathway.
Collapse
MESH Headings
- Animals
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/drug effects
- Calcineurin/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Disease Models, Animal
- NF-kappa B/metabolism
- Down-Regulation
- Dantrolene/pharmacology
- Male
- Calcineurin Inhibitors/pharmacology
- NFATC Transcription Factors/metabolism
- Cells, Cultured
- Cardiomegaly/metabolism
- Cardiomegaly/prevention & control
- Cardiomegaly/pathology
- Cardiomegaly/drug therapy
- Rats, Sprague-Dawley
- Rats
- Hypertrophy, Left Ventricular/prevention & control
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
Collapse
Affiliation(s)
- Yi Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Shuai Li
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xueyan Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Daoyuan Si
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Fenghua Yang
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Huan Sun
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China.
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China.
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China.
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China.
| |
Collapse
|
5
|
Alim CC, Ko CY, Mira Hernandez J, Shen EY, Baidar S, Chen‐Izu Y, Bers DM, Bossuyt J. Nitrosylation of cardiac CaMKII at Cys290 mediates mechanical afterload-induced increases in Ca 2+ transient and Ca 2+ sparks. J Physiol 2022; 600:4865-4879. [PMID: 36227145 PMCID: PMC9827875 DOI: 10.1113/jp283427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/30/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiac mechanical afterload induces an intrinsic autoregulatory increase in myocyte Ca2+ dynamics and contractility to enhance contraction (known as the Anrep effect or slow force response). Our prior work has implicated both nitric oxide (NO) produced by NO synthase 1 (NOS1) and calcium/calmodulin-dependent protein kinase II (CaMKII) activity as required mediators of this form of mechano-chemo-transduction. To test whether a single S-nitrosylation site on CaMKIIδ (Cys290) mediates enhanced sarcoplasmic reticulum Ca2+ leak and afterload-induced increases in sarcoplasmic reticulum (SR) Ca2+ uptake and release, we created a novel CRISPR-based CaMKIIδ knock-in (KI) mouse with a Cys to Ala mutation at C290. These CaMKIIδ-C290A-KI mice exhibited normal cardiac morphometry and function, as well as basal myocyte Ca2+ transients (CaTs) and β-adrenergic responses. However, the NO donor S-nitrosoglutathione caused an acute increased Ca2+ spark frequency in wild-type (WT) myocytes that was absent in the CaMKIIδ-C290A-KI myocytes. Using our cell-in-gel system to exert multiaxial three-dimensional mechanical afterload on myocytes during contraction, we found that WT myocytes exhibited an afterload-induced increase in Ca2+ sparks and Ca2+ transient amplitude and rate of decline. These afterload-induced effects were prevented in both cardiac-specific CaMKIIδ knockout and point mutant CaMKIIδ-C290A-KI myocytes. We conclude that CaMKIIδ activation by S-nitrosylation at the C290 site is essential in mediating the intrinsic afterload-induced enhancement of myocyte SR Ca2+ uptake, release and Ca2+ transient amplitude (the Anrep effect). The data also indicate that NOS1 activation is upstream of S-nitrosylation at C290 of CaMKII, and that this molecular mechano-chemo-transduction pathway is beneficial in allowing the heart to increase contractility to limit the reduction in stroke volume when aortic pressure (afterload) is elevated. KEY POINTS: A novel CRISPR-based CaMKIIδ knock-in mouse was created in which kinase activation by S-nitrosylation at Cys290 (C290A) is prevented. How afterload affects Ca2+ signalling was measured in cardiac myocytes that were embedded in a hydrogel that imposes a three-dimensional afterload. This mechanical afterload induced an increase in Ca2+ transient amplitude and decay in wild-type myocytes, but not in cardiac-specific CaMKIIδ knockout or C290A knock-in myocytes. The CaMKIIδ-C290 S-nitrosylation site is essential for the afterload-induced enhancement of Ca2+ transient amplitude and Ca2+ sparks.
Collapse
Affiliation(s)
- Chidera C. Alim
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA
| | | | - Juliana Mira Hernandez
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA,Research Group in Veterinary MedicineSchool of Veterinary MedicineUniversity Corporation LasallistaCaldasAntioquiaColombia
| | - Erin Y. Shen
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA
| | - Sonya Baidar
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA
| | - Ye Chen‐Izu
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA,Department of Biomedical EngineeringUniversity of CaliforniaDavisCAUSA,Department of Internal Medicine/CardiologyUniversity of CaliforniaDavisCAUSA
| | - Donald M. Bers
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA
| | - Julie Bossuyt
- Department of PharmacologyUniversity of CaliforniaDavisCAUSA
| |
Collapse
|
6
|
Rocco-Machado N, Lai L, Kim G, He Y, Luczak ED, Anderson ME, Levine RL. Oxidative stress–induced autonomous activation of the calcium/calmodulin-dependent kinase II involves disulfide formation in the regulatory domain. J Biol Chem 2022; 298:102579. [PMID: 36220393 PMCID: PMC9643438 DOI: 10.1016/j.jbc.2022.102579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase II δ (CaMKIIδ) has a pivotal role in cardiac signaling. Constitutive and deleterious CaMKII “autonomous” activation is induced by oxidative stress, and the previously reported mechanism involves oxidation of methionine residues in the regulatory domain. Here, we demonstrate that covalent oxidation leads to a disulfide bond with Cys273 in the regulatory domain causing autonomous activity. Autonomous activation was induced by treating CaMKII with diamide or histamine chloramine, two thiol-oxidizing agents. Autonomy was reversed when the protein was incubated with DTT or thioredoxin to reduce disulfide bonds. Tryptic mapping of the activated CaMKII revealed formation of a disulfide between Cys273 and Cys290 in the regulatory domain. We determined the apparent pKa of those Cys and found that Cys273 had a low pKa while that of Cys290 was elevated. The low pKa of Cys273 facilitates oxidation of its thiol to the sulfenic acid at physiological pH. The reactive sulfenic acid then attacks the thiol of Cys290 to form the disulfide. The previously reported CaMKII mutant in which methionine residues 281 and 282 were mutated to valine (MMVV) protects mice and flies from cardiac decompensation induced by oxidative stress. Our initial hypothesis was that the MMVV mutant underwent a conformational change that prevented disulfide formation and autonomous activation. However, we found that the thiol-oxidizing agents induced autonomy in the MMVV mutant and that the mutant undergoes rapid degradation by the cell, potentially preventing accumulation of the injurious autonomous form. Together, our results highlight additional mechanistic details of CaMKII autonomous activation.
Collapse
Affiliation(s)
- Nathália Rocco-Machado
- Laboratory of Biochemistry, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Lo Lai
- Laboratory of Biochemistry, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Geumsoo Kim
- Laboratory of Biochemistry, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Yi He
- Fermentation Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth D Luczak
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark E Anderson
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Physiology and Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rodney L Levine
- Laboratory of Biochemistry, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA.
| |
Collapse
|
7
|
Costa BM, Mengal V, Brasil GA, Peluso AA, Treebak JT, Endlich PW, de Almeida SA, de Abreu GR. Ellagic Acid Prevents Myocardial Infarction-induced Left Ventricular Diastolic Dysfunction in Ovariectomized Rats. J Nutr Biochem 2022; 105:108990. [PMID: 35331902 DOI: 10.1016/j.jnutbio.2022.108990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 11/18/2021] [Accepted: 02/22/2022] [Indexed: 12/07/2022]
Abstract
Estrogen deficiency is associated with increased oxidative stress, which can contribute to left ventricular diastolic dysfunction (LVDD). We hypothesized that oral treatment with ellagic acid (EA), a potent and natural antioxidant compound, can improve MI-induced LVDD in ovariectomized rats, by reducing the formation of reactive oxygen species (ROS). Ovariectomized rats MI-induced LVDD followed by treatment with vehicle (DD) or EA (DD+EA) for 4 weeks. Non-LVDD-induced rats treated with vehicle (S) or EA (S+EA) were used as controls. Left ventricular systolic pressure: LVSP; left ventricular end-diastolic pressure: LVEDP; maximum rate of pressure rise: +dP/dt and fall: -dP/dt) were evaluated in all animals after treatment. Left ventricle superoxide anion formation was quantified in situ by fluorescence. Phospho-CAMKII, SOD2, catalase and gp91-phox abundances were evaluated by Western blot analyses. SOD and catalase activities were measured by spectrophotometry. The results showed that the LVEDP was significantly increased in both DD and DD+EA groups compared to S and S+EA. However, LVEDP in the DD+EA group was significantly decreased compared to DD, indicating an EA-mediated effect. In the DD group, superoxide production and gp91-phox protein abundance were increased while SOD2 abundance was decreased when compared to the S and S+EA groups. An increase in SOD activity was also observed in the DD+EA group. EA treatment reduced CaMKII phosphorylation in the DD+EA group compared to the DD. We concluded that EA treatment attenuated diastolic dysfunction in our experimental model, via reduction of ROS and CaMKII activity, indicating EA as a promising natural therapeutic option for cardiac dysfunction.
Collapse
Affiliation(s)
- Bruno Maia Costa
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Vinícius Mengal
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, ES, Brazil
| | | | - Antônio Augusto Peluso
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Patrick Wander Endlich
- Faculdade de Medicina do Mucuri, Multicentric Post-Graduate Program in Physiological Sciences, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Teófilo Otoni, MG, Brazil
| | - Simone Alves de Almeida
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, ES, Brazil.
| | - Gláucia Rodrigues de Abreu
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
8
|
An N, Zhang G, Li Y, Yuan C, Yang F, Zhang L, Gao Y, Xing Y. Promising Antioxidative Effect of Berberine in Cardiovascular Diseases. Front Pharmacol 2022; 13:865353. [PMID: 35321323 PMCID: PMC8936808 DOI: 10.3389/fphar.2022.865353] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Berberine (BBR), an important quaternary benzylisoquinoline alkaloid, has been used in Chinese traditional medicine for over 3,000 years. BBR has been shown in both traditional and modern medicine to have a wide range of pharmacological actions, including hypoglycemic, hypolipidemic, anti-obesity, hepatoprotective, anti-inflammatory, and antioxidant activities. The unregulated reaction chain induced by oxidative stress as a crucial mechanism result in myocardial damage, which is involved in the pathogenesis and progression of many cardiovascular diseases (CVDs). Numerous researches have established that BBR protects myocardium and may be beneficial in the treatment of CVDs. Given that the pivotal role of oxidative stress in CVDs, the pharmacological effects of BBR in the treatment and/or management of CVDs have strongly attracted the attention of scholars. Therefore, this review sums up the prevention and treatment mechanisms of BBR in CVDs from in vitro, in vivo, and finally to the clinical field trials timely. We summarized the antioxidant stress of BBR in the management of coronary atherosclerosis and myocardial ischemia/reperfusion; it also analyzes the pathogenesis of oxidative stress in arrhythmia and heart failure and the therapeutic effects of BBR. In short, BBR is a hopeful drug candidate for the treatment of CVDs, which can intervene in the process of CVDs from multiple angles and different aspects. Therefore, if we want to apply it to the clinic on a large scale, more comprehensive, intensive, and detailed researches are needed to be carried out to clarify the molecular mechanism and targets of BBR.
Collapse
Affiliation(s)
- Na An
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guoxia Zhang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingjian Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chao Yuan
- Dezhou Second People’s Hospital, Dezhou, China
| | - Fan Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lijing Zhang
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Gao R, Li X, Gao H, Zhao K, Liu X, Liu J, Wang Q, Zhu Y, Chen H, Xiang S, Zhan Y, Yin R, Yu M, Ning H, Yang X, Li C. Protein phosphatase 2A catalytic subunit β suppresses PMA/ionomycin-induced T-cell activation by negatively regulating PI3K/Akt signaling. FEBS J 2022; 289:4518-4535. [PMID: 35068054 DOI: 10.1111/febs.16370] [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: 08/19/2021] [Revised: 12/16/2021] [Accepted: 01/20/2022] [Indexed: 01/07/2023]
Abstract
The precise regulation of the T-cell activation process is critical for overall immune homeostasis. Although protein phosphatase 2A (PP2A) is required for T-cell development and function, the role of PPP2CB, which is the catalytic subunit β isoform of PP2A, remains unknown. In the present study, using a T cell-specific knockout mouse of PPP2CB (PPP2CBfl/fl Lck-Cre+ ), we demonstrated that PPP2CB was dispensable for T-cell development in the thymus and peripheral lymphoid organs. Furthermore, PPP2CB deletion did not affect T-cell receptor (TCR)-induced T-cell activation or cytokine-induced T-cell responses; however, it specifically enhanced phorbol myristate acetate (PMA) plus ionomycin-induced T-cell activation with increased cellular proliferation, elevated CD69 and CD25 expression, and enhanced cytokine production (inteferon-γ, interleukin-2 and tumor necrosis factor). Mechanistic analyses suggested that the PPP2CB deletion enhanced activation of the phosphoinositide 3-kinase/Akt signaling pathway and Ca2+ flux following stimulation with PMA plus ionomycin. Moreover, the specific PI3K inhibitor rescued the augmented cell activation in PPP2CB-deficient T cells. Using mass spectrometry-based phospho-peptide analysis, we identified potential substrates of PPP2CB during PMA plus ionomycin-induced T-cell activation. Collectively, our study provides evidence of the specific role of PPP2CB in controlling PMA plus ionomycin-induced T-cell activation.
Collapse
Affiliation(s)
- Rui Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Xin Li
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Huiying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Xian Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Jinfang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Qi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yaxin Zhu
- School of Life Sciences, Hebei University, Baoding, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Shensi Xiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Yiqun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Ronghua Yin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Hongmei Ning
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| | - Changyan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, China
| |
Collapse
|
10
|
D’Aloia A, Arrigoni E, Costa B, Berruti G, Martegani E, Sacco E, Ceriani M. RalGPS2 Interacts with Akt and PDK1 Promoting Tunneling Nanotubes Formation in Bladder Cancer and Kidney Cells Microenvironment. Cancers (Basel) 2021; 13:cancers13246330. [PMID: 34944949 PMCID: PMC8699646 DOI: 10.3390/cancers13246330] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Cell-to-cell communication in the tumor microenvironment is a crucial process to orchestrate the different components of the tumoral infrastructure. Among the mechanisms of cellular interplay in cancer cells, tunneling nanotubes (TNTs) are dynamic connections that play an important role. The mechanism of the formation of TNTs among cells and the molecules involved in the process remain to be elucidated. In this study, we analyze several bladder cancer cell lines, representative of tumors at different stages and grades. We demonstrate that TNTs are formed only by mid or high-stage cell lines that show muscle-invasive properties and that they actively transport mitochondria and proteins. The formation of TNTs is triggered by stressful conditions and starts with the assembly of a specific multimolecular complex. In this study, we characterize some of the protein components of the TNTs complex, as they are potential novel molecular targets for future therapies aimed at counteracting tumor progression. Abstract RalGPS2 is a Ras-independent Guanine Nucleotide Exchange Factor for RalA GTPase that is involved in several cellular processes, including cytoskeletal organization. Previously, we demonstrated that RalGPS2 also plays a role in the formation of tunneling nanotubes (TNTs) in bladder cancer 5637 cells. In particular, TNTs are a novel mechanism of cell–cell communication in the tumor microenvironment, playing a central role in cancer progression and metastasis formation. However, the molecular mechanisms involved in TNTs formation still need to be fully elucidated. Here we demonstrate that mid and high-stage bladder cancer cell lines have functional TNTs, which can transfer mitochondria. Moreover, using confocal fluorescence time-lapse microscopy, we show in 5637 cells that TNTs mediate the trafficking of RalA protein and transmembrane MHC class III protein leukocyte-specific transcript 1 (LST1). Furthermore, we show that RalGPS2 is essential for nanotubes generation, and stress conditions boost its expression both in 5637 and HEK293 cell lines. Finally, we prove that RalGPS2 interacts with Akt and PDK1, in addition to LST1 and RalA, leading to the formation of a complex that promotes nanotubes formation. In conclusion, our findings suggest that in the tumor microenvironment, RalGPS2 orchestrates the assembly of multimolecular complexes that drive the formation of TNTs.
Collapse
Affiliation(s)
- Alessia D’Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Edoardo Arrigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Giovanna Berruti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy;
| | - Enzo Martegani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milano, Italy
- Correspondence: ; Tel.: +39-0264483544
| |
Collapse
|
11
|
Xu D, Murakoshi N, Tajiri K, Duo F, Okabe Y, Murakata Y, Yuan Z, Li S, Aonuma K, Song Z, Shimoda Y, Mori H, Sato A, Nogami A, Aonuma K, Ieda M. Xanthine oxidase inhibitor febuxostat reduces atrial fibrillation susceptibility by inhibition of oxidized CaMKII in Dahl salt-sensitive rats. Clin Sci (Lond) 2021; 135:2409-2422. [PMID: 34386810 DOI: 10.1042/cs20210405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.
Collapse
Affiliation(s)
- DongZhu Xu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Feng Duo
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuta Okabe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zixun Yuan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zonghu Song
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuzuno Shimoda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
12
|
Santos-Miranda A, Costa AD, Joviano-Santos JV, Rhana P, Bruno AS, Rocha P, Cau SB, Vieira LQ, Cruz JS, Roman-Campos D. Inhibition of calcium/calmodulin (Ca 2+ /CaM)-Calcium/calmodulin-dependent protein kinase II (CaMKII) axis reduces in vitro and ex vivo arrhythmias in experimental Chagas disease. FASEB J 2021; 35:e21901. [PMID: 34569665 DOI: 10.1096/fj.202101060r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/11/2022]
Abstract
Chagasic cardiomyopathy (CCC) is one of the main causes of heart failure and sudden death in Latin America. To date, there is no available medication to prevent or reverse the onset of cardiac symptoms. CCC occurs in a scenario of disrupted calcium dynamics and enhanced oxidative stress, which combined, may favor the hyper activation of calcium/calmodulin (Ca2+ /CaM)-calcium/calmodulin-dependent protein kinase II (CaMKII) (Ca2+ /CaM-CaMKII) pathway, which is fundamental for heart physiology and it is implicated in other cardiac diseases. Here, we evaluated the association between Ca2+ /CaM-CaMKII in the electro-mechanical (dys)function of the heart in the early stage of chronic experimental Trypanosoma cruzi infection. We observed that in vitro and ex vivo inhibition of Ca2+ /CaM-CaMKII reversed the arrhythmic profile of isolated hearts and isolated left-ventricles cardiomyocytes. The benefits of the limited Ca2+ /CaM-CaMKII activation to cardiomyocytes' electrical properties are partially related to the restoration of Ca2+ dynamics in a damaged cellular environment created after T. cruzi infection. Moreover, Ca2+ /CaM-CaMKII inhibition prevented the onset of arrhythmic contractions on isolated heart preparations of chagasic mice and restored the responsiveness to the increase in the left-ventricle pre-load. Taken together, our data provide the first experimental evidence for the potential of targeting Ca2+ /CaM-CaMKII pathway as a novel therapeutic target to treat CCC.
Collapse
Affiliation(s)
| | - Alexandre D Costa
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Paula Rhana
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre Santos Bruno
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Peter Rocha
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stefany Bruno Cau
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leda Q Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Aravind P, Bulbule SR, Hemalatha N, Babu R, Devaraju K. Elevation of gene expression of calcineurin, calmodulin and calsyntenin in oxidative stress induced PC12 cells. Genes Dis 2021; 8:87-93. [PMID: 33569517 PMCID: PMC7859428 DOI: 10.1016/j.gendis.2019.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/01/2019] [Accepted: 09/01/2019] [Indexed: 12/30/2022] Open
Abstract
In normal physiological conditions, reactive oxygen and nitrogen species are used as important signaling molecules in the cell. However, in excess it causes the disruption of cell resulting in their death. Oxidative stress causes influx in intracellular calcium levels leading to higher concentrations of calcium in the cell. This accelerated calcium affects both the mitochondria and nuclei leading to excitotoxicity in neurons. Intracellular calcium levels are controlled by voltage dependent calcium channels located in the plasma membrane, calcium stores like endoplasmic/sarcoplasmic reticulum and majorly by calcium binding proteins. Our study was aimed at analyzing the gene expression of major calcium binding proteins namely calcineurin, calmodulin, calreticulin, synaptotagamin and calsyntenin in stress induced PC 12 cells. Rotenone (1 μM), Peroxynitrite (10 μM), H2O2 (100 μM) and High glucose (33 mM) were used to induce oxidative stress in PC12 cells. Results obtained from the study suggest that calcineurin, calmodulin and calsyntenin gene expression were enhanced compared to the control due to oxidative stress. However, synaptotagmin and calreticulin gene expression were down regulated. Further, Akt protein expression (stress marker) was enhanced in PC12 cells with all other stress inducers except in hyperglycemic condition.
Collapse
Affiliation(s)
- P. Aravind
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| | - Sarojini R. Bulbule
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| | - N. Hemalatha
- Department of Biochemistry and Nutrition, CFTRI, V V Moholla, Mysore, 570 020, India
| | - R.L. Babu
- Department of Bioinformatics and Biotechnology, Akkamahadevi Women's University, Vijayapura, Karnataka, 586 108, India
| | - K.S. Devaraju
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| |
Collapse
|
14
|
Takata T, Araki S, Tsuchiya Y, Watanabe Y. Persulfide Signaling in Stress-Initiated Calmodulin Kinase Response. Antioxid Redox Signal 2020; 33:1308-1319. [PMID: 32460522 DOI: 10.1089/ars.2020.8138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Significance: Calcium ion (Ca2+)/calmodulin (CaM)-dependent protein kinases (CaMKs) are activated by phosphorylation of a crucial threonine residue either by itself (CaMKII) or by upstream kinases, CaMK kinases (CaMKKs) (CaMKI and CaMKIV). CaMKs, present in most mammalian tissues, can phosphorylate many downstream targets, thereby regulating numerous cellular functions. Recent Advances: Aside from canonical post-translational modifications, cysteine-based redox switches in CaMKs affect their enzyme activities. In addition to reactive oxygen species (ROS) and reactive nitrogen species (RNS), reactive sulfur species (RSS) are also recognized as key signaling molecules, regulating protein function through polysulfidation, formation of polysulfides [-S-(S)n-H] on their reactive cysteine residues. To comprehend the biological significance of RSS signaling-related CaMK regulation, here we introduce a novel concept defining CaMKs as RSS targets in stress responses. The stress responses include an irreversible electrophile attack for CaMKI, inflammation for CaMKII, and endoplasmic reticulum stress for CaMKIV. Critical Issues: Development of various human diseases is associated with increased ROS, RNS, and RSS generation. Therefore, depending on specific pathophysiology, RSS could have very particular effects on CaMK functions. Future Directions: How multiple sources and mutual reactions of ROS, RNS, and RSS are coordinated is obscure. Elucidating the mechanisms through applications of enzymology, chemical biology, and mass spectrometry enables to uncover the complexities of redox regulation of CaMK cascades.
Collapse
Affiliation(s)
- Tsuyoshi Takata
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan.,Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoma Araki
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
15
|
Oxidative Stress Orchestrates MAPK and Nitric-Oxide Synthase Signal. Int J Mol Sci 2020; 21:ijms21228750. [PMID: 33228180 PMCID: PMC7699490 DOI: 10.3390/ijms21228750] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) are not only harmful to cell survival but also essential to cell signaling through cysteine-based redox switches. In fact, ROS triggers the potential activation of mitogen-activated protein kinases (MAPKs). The 90 kDa ribosomal S6 kinase 1 (RSK1), one of the downstream mediators of the MAPK pathway, is implicated in various cellular processes through phosphorylating different substrates. As such, RSK1 associates with and phosphorylates neuronal nitric oxide (NO) synthase (nNOS) at Ser847, leading to a decrease in NO generation. In addition, the RSK1 activity is sensitive to inhibition by reversible cysteine-based redox modification of its Cys223 during oxidative stress. Aside from oxidative stress, nitrosative stress also contributes to cysteine-based redox modification. Thus, the protein kinases such as Ca2+/calmodulin (CaM)-dependent protein kinase I (CaMKI) and II (CaMKII) that phosphorylate nNOS could be potentially regulated by cysteine-based redox modification. In this review, we focus on the role of post-translational modifications in regulating nNOS and nNOS-phosphorylating protein kinases and communication among themselves.
Collapse
|
16
|
Zhang X, Griepentrog JE, Zou B, Xu L, Cyr AR, Chambers LM, Zuckerbraun BS, Shiva S, Rosengart MR. CaMKIV regulates mitochondrial dynamics during sepsis. Cell Calcium 2020; 92:102286. [PMID: 32932146 DOI: 10.1016/j.ceca.2020.102286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Sepsis and shock states impose mitochondrial stress, and in response, adaptive mechanisms such as fission, fusion and mitophagy are induced to eliminate damaged portions of or entire dysfunctional mitochondria. The mechanisms underlying these events are being elucidated; yet a direct link between loss of mitochondrial membrane potential ΔΨm and the initiation of fission, fusion and mitophagy remains to be well characterized. The direct association between the magnitude of the ΔΨm and the capacity for mitochondria to buffer Ca2+ renders Ca2+ uniquely suited as the signal engaging these mechanisms in circumstances of mitochondrial stress that lower the ΔΨm. Herein, we show that the calcium/calmodulin-dependent protein kinase (CaMK) IV mediates an adaptive slowing in oxidative respiration that minimizes oxidative stress in the kidneys of mice subjected to either cecal ligation and puncture (CLP) sepsis or endotoxemia. CaMKIV shifts the balance towards mitochondrial fission and away from fusion by 1) directly phosphorylating an activating Serine616 on the fission protein DRP1 and 2) reducing the expression of the fusion proteins Mfn1/2 and OPA-1. CaMKIV, through its function as a direct PINK1 kinase and regulator of Parkin expression, also enables mitophagy. These data support that CaMKIV serves as a keystone linking mitochondrial stress with the adaptive mechanisms of mitochondrial fission, fusion and mitophagy that mitigate oxidative stress in the kidneys of mice responding to sepsis.
Collapse
Affiliation(s)
- Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - John E Griepentrog
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Baobo Zou
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States; Department of Emergency, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Anthony R Cyr
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lauran M Chambers
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
17
|
Chowdhury AR, Zielonka J, Kalyanaraman B, Hartley RC, Murphy MP, Avadhani NG. Mitochondria-targeted paraquat and metformin mediate ROS production to induce multiple pathways of retrograde signaling: A dose-dependent phenomenon. Redox Biol 2020; 36:101606. [PMID: 32604037 PMCID: PMC7327929 DOI: 10.1016/j.redox.2020.101606] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/11/2020] [Indexed: 01/12/2023] Open
Abstract
The mitochondrial electron transport chain is a major source of reactive oxygen species (ROS) and is also a target of ROS, with an implied role in the stabilization of hypoxia-inducible factor (HIF) and induction of the AMPK pathway. Here we used varying doses of two agents, Mito-Paraquat and Mito-Metformin, that have been conjugated to cationic triphenylphosphonium (TPP+) moiety to selectively target them to the mitochondrial matrix compartment, thereby resulting in the site-specific generation of ROS within mitochondria. These agents primarily induce superoxide (O2•-) production by acting on complex I. In Raw264.7 macrophages, C2C12 skeletal myocytes, and HCT116 adenocarcinoma cells, we show that mitochondria-targeted oxidants can induce ROS (O2•- and H2O2). In all three cell lines tested, the mitochondria-targeted agents disrupted membrane potential and activated calcineurin and the Cn-dependent retrograde signaling pathway. Hypoxic culture conditions also induced Cn activation and HIF1α activation in a temporally regulated manner, with the former appearing at shorter exposure times. Together, our results indicate that mitochondrial oxidant-induced retrograde signaling is driven by disruption of membrane potential and activation of Ca2+/Cn pathway and is independent of ROS-induced HIF1α or AMPK pathways.
Collapse
Affiliation(s)
- Anindya Roy Chowdhury
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacek Zielonka
- Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Balaraman Kalyanaraman
- Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Michael P Murphy
- MRC-Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 OXY, UK
| | - Narayan G Avadhani
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Yang X, An N, Zhong C, Guan M, Jiang Y, Li X, Zhang H, Wang L, Ruan Y, Gao Y, Liu N, Shang H, Xing Y. Enhanced cardiomyocyte reactive oxygen species signaling promotes ibrutinib-induced atrial fibrillation. Redox Biol 2020; 30:101432. [PMID: 31986467 PMCID: PMC6994714 DOI: 10.1016/j.redox.2020.101432] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/27/2019] [Accepted: 01/12/2020] [Indexed: 12/12/2022] Open
Abstract
Atrial fibrillation (AF) occurs in up to 11% of cancer patients treated with ibrutinib. The pathophysiology of ibrutinib promoted AF is complicated, as there are multiple interactions involved; the detailed molecular mechanisms underlying this are still unclear. Here, we aimed to determine the electrophysiological and molecular mechanisms of burst-pacing-induced AF in ibrutinib-treated mice. The results indicated differentially expressed proteins in ibrutinib-treated mice, identified through proteomic analysis, were found to play a role in oxidative stress-related pathways. Finally, treatment with an inhibitor of NADPH oxidase (NOX) prevented and reversed AF development in ibrutinib-treated mice. It was showed that the related protein expression of reactive oxygen species (ROS) in the ibrutinib group was significantly increased, including NOX2, NOX4, p22-phox, XO and TGF-β protein expression. It was interesting that ibrutinib group also significantly increased the expression of ox-CaMKII, p-CaMKII (Thr-286) and p-RyR2 (Ser2814), causing enhanced abnormal sarcoplasmic reticulum (SR) Ca2+ release and mitochondrial structures, as well as atrial fibrosis and atrial hypertrophy in ibrutinib-treated mice, and apocynin reduced the expression of these proteins. Ibrutinib-treated mice were also more likely to develop AF, and AF occurred over longer periods. In conclusion, our study has established a pathophysiological role for ROS signaling in atrial cardiomyocytes, and it may be that ox-CaMKII and p-CaMKII (Thr-286) are activated by ROS to increase AF susceptibility following ibrutinib treatment. We have also identified the inhibition of NOX as a potential novel AF therapy approach.
Collapse
Affiliation(s)
- Xinyu Yang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Changming Zhong
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Manke Guan
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yuchen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xinye Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Hanlai Zhang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Liqin Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yanfei Ruan
- Department of Cardiology, Beijing An Zhen Hospital of the Capital University of Medical Sciences, Beijing, 100853, PR China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Nian Liu
- Department of Cardiology, Beijing An Zhen Hospital of the Capital University of Medical Sciences, Beijing, 100853, PR China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
19
|
Mancini A, Fuvuzzi AMR, Bruno C, Nicolazzi MA, Vergani E, Ciferri N, Silvestrini A, Meucci E, Nicolotti N, D'Assante R, Cittadini A. Anabolic Hormone Deficiencies in Heart Failure with Reduced or Preserved Ejection Fraction and Correlation with Plasma Total Antioxidant Capacity. Int J Endocrinol 2020; 2020:5798146. [PMID: 32411227 PMCID: PMC7199626 DOI: 10.1155/2020/5798146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND While anabolic hormone deficit is a common finding in heart failure with reduced ejection fraction (HFrEF), few data are available in heart failure with preserved ejection fraction (HFpEF). METHODS Blood samples were collected for metabolic (total cholesterol, HDL cholesterol, LDL cholesterol, creatinine, and glucose) and hormonal (IGF-1, DHEA-S, TSH, fT3, fT4, and T) determination, comparing 30 patients with HFpEF and 20 patients with HFrEF. Total antioxidant capacity was evaluated by using the spectrophotometric method using the latency time in the appearance of the radical species of a chromogen (LAG, sec) as a parameter proportional to antioxidant content of the sample. Echocardiographic parameters were also assessed in the two groups. RESULTS A high prevalence of testosterone (32% in HFrEF and 72% in HFpEF, p < 0.05) and DHEA-S deficiencies was observed in HFpEF patients. Echocardiographic parameters did not correlate with hormone values. A significant direct correlation between T (r 2 = 0.25, p < 0.05) and DHEA-S (r 2 = 0.19, p < 0.05) with LAG was observed only in HFpEF. CONCLUSION Anabolic hormone deficiency is clearly shown in HFpEF, as already known in HFrEF. Although longitudinal studies are required to confirm the prognostic value of this observation, our data suggest different mechanisms in modulating antioxidants in the two conditions, with possible therapeutic implications.
Collapse
Affiliation(s)
- Antonio Mancini
- Operative Unit of Endocrinology, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angela Maria Rita Fuvuzzi
- Operative Unit of Internal Medicine and Vascular Diseases, Division of Internal Medicine and Cardiovascular Diseases, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmine Bruno
- Operative Unit of Endocrinology, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Anna Nicolazzi
- Operative Unit of Internal Medicine and Vascular Diseases, Division of Internal Medicine and Cardiovascular Diseases, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Edoardo Vergani
- Operative Unit of Endocrinology, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nunzia Ciferri
- Operative Unit of Endocrinology, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Silvestrini
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisabetta Meucci
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nicola Nicolotti
- Medical Management, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
| |
Collapse
|
20
|
Alli-Oluwafuyi AM, Luis PB, Nakashima F, Giménez-Bastida JA, Presley SH, Duvernay MT, Iwalewa EO, Schneider C. Curcumin induces secretion of glucagon-like peptide-1 through an oxidation-dependent mechanism. Biochimie 2019; 165:250-257. [PMID: 31470039 DOI: 10.1016/j.biochi.2019.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/24/2019] [Indexed: 10/26/2022]
Abstract
Curcumin shows antiglycemic effects in animals. Curcumin is chemically unstable at physiological pH, and its oxidative degradation products were shown to contribute to its anti-inflammatory effects. Since the degradation products may also contribute to other effects, we analyzed their role in the antiglycemic activity of curcumin. We quantified curcumin-induced release of glucagon-like peptide 1 (GLP-1) from mouse STC-1 cells that represent enteroendocrine L-cells as a major source of this anti-diabetic hormone. Curcumin induced secretion of GLP-1 in a dose-dependent manner. Two chemically stable analogues of curcumin that do not readily undergo degradation, were less active while two unstable analogues were active secretagogues. Chromatographically isolated spiroepoxide, an unstable oxidative metabolite of curcumin with anti-inflammatory activity, also induced secretion of GLP-1. Stable compounds like the final oxidative metabolite bicyclopentadione, and the major plasma metabolite, curcumin-glucuronide, were inactive. GLP-1 secretion induced by curcumin and its oxidative degradation products was associated with activation of PKC, ERK, and CaM kinase II. Since activity largely correlated with instability of curcumin and the analogues, we tested the extent of covalent binding to proteins in STC-1 cells and found it occurred with similar affinity as N-ethylmaleimide, indicating covalent binding occurred with nucleophilic cysteine residues. These results suggest that oxidative metabolites of curcumin are involved in the antiglycemic effects of curcumin. Our findings support the hypothesis that curcumin functions as a pro-drug requiring oxidative activation to reveal its bioactive metabolites that act by binding to target proteins thereby causing a change in function.
Collapse
Affiliation(s)
- Abdul-Musawwir Alli-Oluwafuyi
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA; Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria
| | - Paula B Luis
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Fumie Nakashima
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Juan A Giménez-Bastida
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Sai Han Presley
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Matthew T Duvernay
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Ezekiel O Iwalewa
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Claus Schneider
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA.
| |
Collapse
|
21
|
Myocardial death and dysfunction after ischemia-reperfusion injury require CaMKIIδ oxidation. Sci Rep 2019; 9:9291. [PMID: 31243295 PMCID: PMC6595001 DOI: 10.1038/s41598-019-45743-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023] Open
Abstract
Reactive oxygen species (ROS) contribute to myocardial death during ischemia-reperfusion (I/R) injury, but detailed knowledge of molecular pathways connecting ROS to cardiac injury is lacking. Activation of the Ca2+/calmodulin-dependent protein kinase II (CaMKIIδ) is implicated in myocardial death, and CaMKII can be activated by ROS (ox-CaMKII) through oxidation of regulatory domain methionines (Met281/282). We examined I/R injury in mice where CaMKIIδ was made resistant to ROS activation by knock-in replacement of regulatory domain methionines with valines (MMVV). We found reduced myocardial death, and improved left ventricular function 24 hours after I/R injury in MMVV in vivo and in vitro compared to WT controls. Loss of ATP sensitive K+ channel (KATP) current contributes to I/R injury, and CaMKII promotes sequestration of KATP from myocardial cell membranes. KATP current density was significantly reduced by H2O2 in WT ventricular myocytes, but not in MMVV, showing ox-CaMKII decreases KATP availability. Taken together, these findings support a view that ox-CaMKII and KATP are components of a signaling axis promoting I/R injury by ROS.
Collapse
|
22
|
Takata T, Kimura J, Ihara H, Hatano N, Tsuchiya Y, Watanabe Y. Redox regulation of Ca 2+/calmodulin-dependent protein kinase IV via oxidation of its active-site cysteine residue. Free Radic Biol Med 2019; 130:99-106. [PMID: 30394289 DOI: 10.1016/j.freeradbiomed.2018.10.440] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 09/15/2018] [Accepted: 10/24/2018] [Indexed: 11/15/2022]
Abstract
We have recently reported that Ca2+/calmodulin (CaM)-dependent protein kinase IV (CaMKIV) is inactivated by reactive sulfur species via polysulfidation of the active-site Cys residue. Here, we show that hydrogen peroxide (H2O2) limit CaMKIV activity at the same active-site Cys residue through oxidation and downstream signaling in cells. CaMKIV is phosphorylated at Thr196 by its upstream CaMK kinase (CaMKK), which induces its full activity. In vitro incubation of CaMKIV with H2O2 resulted in reversible inhibition of CaMKK-induced phospho-Thr196 and the consequent inactivation of CaMKIV. In contrast, mutated CaMKIV (C198V) was refractory to the H2O2-induced enzyme inhibition. In transfected cells expressing CaMKIV, Ca2+ ionophore-induced CaMKIV phosphorylation at Thr196 was decreased upon treatment with H2O2, whereas cells expressing mutant CaMKIV (C198V) were resistant to H2O2 treatment. Modification of free thiol with N-ethylmaleimide revealed that Cys198 in CaMKIV is a target for S-oxidation. Additionally, the Ca2+ influx-induced phospho-Thr196 of endogenous CaMKIV was also inhibited upon treatment with H2O2 in Jurkat T-lymphocytes and cerebellar granule cells. Phosphorylation of cyclic AMP response element-binding protein (CREB) at Ser133, which is downstream of CaMKIV, was also decreased upon treatment with H2O2. Thus, our results indicate that oxidation stress regulates cellular function by decreasing the activity of CaMKIV through Cys198 oxidation.
Collapse
Affiliation(s)
- Tsuyoshi Takata
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Jun Kimura
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai 599-8531, Osaka, Japan
| | - Naoya Hatano
- The Integrated Center for Mass Spectrometry, Kobe University, Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
23
|
Song J, Yang R, Yang J, Zhou L. Mitochondrial Dysfunction-Associated Arrhythmogenic Substrates in Diabetes Mellitus. Front Physiol 2018; 9:1670. [PMID: 30574091 PMCID: PMC6291470 DOI: 10.3389/fphys.2018.01670] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022] Open
Abstract
There is increasing evidence that diabetic cardiomyopathy increases the risk of cardiac arrhythmia and sudden cardiac death. While the detailed mechanisms remain incompletely understood, the loss of mitochondrial function, which is often observed in the heart of patients with diabetes, has emerged as a key contributor to the arrhythmogenic substrates. In this mini review, the pathophysiology of mitochondrial dysfunction in diabetes mellitus is explored in detail, followed by descriptions of several mechanisms potentially linking mitochondria to arrhythmogenesis in the context of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jiajia Song
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ruilin Yang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, China
| | - Jing Yang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lufang Zhou
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
24
|
Oliveira MF, Geihs MA, França TFA, Moreira DC, Hermes-Lima M. Is "Preparation for Oxidative Stress" a Case of Physiological Conditioning Hormesis? Front Physiol 2018; 9:945. [PMID: 30116197 PMCID: PMC6082956 DOI: 10.3389/fphys.2018.00945] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/28/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Marcus F Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio A Geihs
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Thiago F A França
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Daniel C Moreira
- Área de Morfologia, Faculdade de Medicina, Universidade de Brasília, Brasilia, Brazil.,Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasilia, Brazil
| | - Marcelo Hermes-Lima
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasilia, Brazil
| |
Collapse
|
25
|
Russo R, Cattaneo F, Lippiello P, Cristiano C, Zurlo F, Castaldo M, Irace C, Borsello T, Santamaria R, Ammendola R, Calignano A, Miniaci MC. Motor coordination and synaptic plasticity deficits are associated with increased cerebellar activity of NADPH oxidase, CAMKII, and PKC at preplaque stage in the TgCRND8 mouse model of Alzheimer's disease. Neurobiol Aging 2018; 68:123-133. [DOI: 10.1016/j.neurobiolaging.2018.02.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/14/2018] [Accepted: 02/24/2018] [Indexed: 10/17/2022]
|
26
|
Bussey CT, Erickson JR. Physiology and pathology of cardiac CaMKII. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
27
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kotla S, Singh NK, Kirchhofer D, Rao GN. Heterodimers of the transcriptional factors NFATc3 and FosB mediate tissue factor expression for 15( S)-hydroxyeicosatetraenoic acid-induced monocyte trafficking. J Biol Chem 2017; 292:14885-14901. [PMID: 28724635 PMCID: PMC5592668 DOI: 10.1074/jbc.m117.804344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Tissue factor (TF) is expressed in vascular and nonvascular tissues and functions in several pathways, including embryonic development, inflammation, and cell migration. Many risk factors for atherosclerosis, including hypertension, diabetes, obesity, and smoking, increase TF expression. To better understand the TF-related mechanisms in atherosclerosis, here we investigated the role of 12/15-lipoxygenase (12/15-LOX) in TF expression. 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), the major product of human 15-LOXs 1 and 2, induced TF expression and activity in a time-dependent manner in the human monocytic cell line THP1. Moreover, TF suppression with neutralizing antibodies blocked 15(S)-HETE-induced monocyte migration. We also found that NADPH- and xanthine oxidase-dependent reactive oxygen species (ROS) production, calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation, and interactions between nuclear factor of activated T cells 3 (NFATc3) and FosB proto-oncogene, AP-1 transcription factor subunit (FosB) are involved in 15(S)-HETE-induced TF expression. Interestingly, NFATc3 first induced the expression of its interaction partner FosB before forming the heterodimeric NFATc3-FosB transcription factor complex, which bound the proximal AP-1 site in the TF gene promoter and activated TF expression. We also observed that macrophages from 12/15-LOX-/- mice exhibit diminished migratory response to monocyte chemotactic protein 1 (MCP-1) and lipopolysaccharide compared with WT mouse macrophages. Similarly, compared with WT macrophages, monocytes from 12/15-LOX-/- mice displayed diminished trafficking, which was rescued by prior treatment with 12(S)-HETE, in a peritonitis model. These observations indicate that 15(S)-HETE-induced monocyte/macrophage migration and trafficking require ROS-mediated CaMKIV activation leading to formation of NFATc3 and FosB heterodimer, which binds and activates the TF promoter.
Collapse
Affiliation(s)
- Sivareddy Kotla
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, California 94080
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| |
Collapse
|
29
|
Jiang X, Wu Z, Lu X, Zhang X, Yu Q, Gan Y, Wu B, Xu Y, Zheng W, Zhang L, Xu F, Ma A, Gan X, Huang S, Yu X, Huang W, Xu R. Activation of CaMKIIγ potentiates T-cell acute lymphoblastic leukemia leukemogenesis via phosphorylating FOXO3a. Oncotarget 2017; 8:75050-75064. [PMID: 29088844 PMCID: PMC5650399 DOI: 10.18632/oncotarget.20504] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/29/2017] [Indexed: 01/07/2023] Open
Abstract
Ca2+/calmodulin–dependent protein kinase II γ (CaMKIIγ) can regulate the proliferation and differentiation of myeloid leukemia cells and accelerate chronic myeloid leukemia blast crisis, but the role of CaMKIIγ in T-cell acute lymphoblastic leukemia (T-ALL) leukemogenesis remains poorly understood. We observed that activated (autophosphorylated) CaMKIIγ was invariably present in T-ALL cell lines and in the majority of primary T-ALL samples. Overexpression of CaMKIIγ enhanced the proliferation, colony formation, in vivo tumorigenesis and increased DNA damage of T-ALL leukemia cells. Furthermore, inhibition of CaMKIIγ activity with a pharmacologic inhibitor, gene knock-out, dominant-negative constructs or enhancement of CaMKIIγ activity by overexpression constructs revealed that the activated CaMKIIγ could phosphorylate FOXO3a. In Jurkat cells, the activated CaMKIIγ phosphorylated FOXO3a via directly or indirectly phosphorylating AKT, excluded FOXO3a from the nucleus and inhibited its transcriptional activity. These results indicate that the activated CaMKIIγ may play a key role in T-ALL leukemogenesis, and targeting CaMKIIγ might be a value approach in the treatment of T-ALL.
Collapse
Affiliation(s)
- Xudong Jiang
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Zhaoxing Wu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Xiaoya Lu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Xuzhao Zhang
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qingfeng Yu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Yichao Gan
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Bowen Wu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Ying Xu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Weiwei Zheng
- Deptartment of Clinical Laboratory of Anhui Provincial Hospital, Anhui Medical University, Hefei 230000, China
| | - Lei Zhang
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Fei Xu
- Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - An Ma
- Zhejiang Academy of Medical Sciences, Hangzhou 310009, China
| | - Xiaoxian Gan
- Zhejiang Academy of Medical Sciences, Hangzhou 310009, China
| | - Silvia Huang
- City of Hope Eugene and Ruth Roberts Summer Student Academy, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Xiaofang Yu
- Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| | - Wendong Huang
- Molecular Oncology Program and Department of Diabetes Complications and Metabolism, Beckman Research Institute, Duarte, CA 91010, USA.,Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Rongzhen Xu
- Department of Hematology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.,Cancer Institute of Zhejiang University, Hangzhou, 310009 China
| |
Collapse
|
30
|
Reactive sulfur species inactivate Ca 2+/calmodulin-dependent protein kinase IV via S-polysulfidation of its active-site cysteine residue. Biochem J 2017. [PMID: 28637792 DOI: 10.1042/bcj20170092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Reactive sulfur species (RSS) modulate protein functions via S-polysulfidation of reactive Cys residues. Here, we report that Ca2+/calmodulin (CaM)-dependent protein kinase IV (CaMKIV) was reversibly inactivated by RSS via polysulfidation of the active-site Cys residue. CaMKIV is phosphorylated at Thr196 by its upstream CaMK kinase (CaMKK), resulting in the induction of its full activity. In vitro incubation of CaMKIV with the exogenous RSS donors Na2S n (n = 2-4) resulted in dose-dependent inhibition of the CaMKK-induced phospho-Thr196 and consequent inactivation of the enzyme activity. Conversely, mutated CaMKIV (C198V) was refractory to the Na2S n -induced enzyme inhibition. A biotin-polyethylene glycol-conjugated maleimide capture assay revealed that Cys198 in CaMKIV represents a target for S-polysulfidation. Furthermore, phosho-Thr196 and CaMKIV activity were inhibited by incubation with cysteine hydropersulfide, a newly identified RSS that is generated from cystine by cystathionine-γ-lyase. In transfected cells expressing CaMKIV, ionomycin-induced CaMKIV phosphorylation at Thr196 was decreased upon treatment with either Na2S4 or the endoplasmic reticulum (ER) stress inducer thapsigargin, whereas cells expressing mutant CaMKIV (C198V) were resistant to this treatment. In addition, the ionomycin-induced phospho-Thr196 of endogenous CaMKIV was also inhibited by treatment either with Na2S4 or thapsigargin in Jurkat T lymphocytes. Taken together, these data define a novel signaling function for intracellular RSS in inhibiting CaMKIV activity via S-polysulfidation of its Cys198 during the response to ER stress.
Collapse
|
31
|
Zhang X, Yuan D, Sun Q, Xu L, Lee E, Lewis AJ, Zuckerbraun BS, Rosengart MR. Calcium/calmodulin-dependent protein kinase regulates the PINK1/Parkin and DJ-1 pathways of mitophagy during sepsis. FASEB J 2017; 31:4382-4395. [PMID: 28615325 DOI: 10.1096/fj.201601096rrr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 06/06/2017] [Indexed: 11/11/2022]
Abstract
During sepsis and shock states, mitochondrial dysfunction occurs. Consequently, adaptive mechanisms, such as fission, fusion, and mitophagy, are induced to eliminate damaged portions or entire dysfunctional mitochondria. The regulatory PINK1/Parkin and DJ-1 pathways are strongly induced by mitochondrial depolarization, although a direct link between loss of mitochondrial membrane potential (ΔΨ) and mitophagy has not been identified. Mitochondria also buffer Ca2+, and their buffering capacity is dependent on ΔΨ Here, we characterize a role for calcium/calmodulin-dependent protein kinase (CaMK) I in the regulation of these mechanisms. Loss of ΔΨ with either pharmacologic depolarization or LPS leads to Ca2+-dependent mitochondrial recruitment and activation of CaMKI that precedes the colocalization of PINK1/Parkin and DJ-1. CaMKI is required and serves as both a PINK1 and Parkin kinase. The mechanisms operate in both immune and nonimmune cells and are induced in in vivo models of endotoxemia, sepsis, and hemorrhagic shock. These data support the idea that CaMKI links mitochondrial stress with the PINK1/Parkin and DJ-1 mechanisms of mitophagy.-Zhang, X., Yuan, D., Sun, Q., Xu, L., Lee, E., Lewis, A. J., Zuckerbraun, B. S., Rosengart, M. R. Calcium/calmodulin-dependent protein kinase regulates the PINK1/Parkin and DJ-1 pathways of mitophagy during sepsis.
Collapse
Affiliation(s)
- Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Du Yuan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qian Sun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Emma Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony J Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
32
|
Cardiac Protection of Valsartan on Juvenile Rats with Heart Failure by Inhibiting Activity of CaMKII via Attenuating Phosphorylation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4150158. [PMID: 28536695 PMCID: PMC5425837 DOI: 10.1155/2017/4150158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/09/2017] [Indexed: 11/18/2022]
Abstract
Background. This study was undertaken to determine relative contributions of phosphorylation and oxidation to the increased activity of calcium/calmodulin-stimulated protein kinase II (CaMKII) in juveniles with cardiac myocyte dysfunction due to increased pressure overload. Methods. Juvenile rats underwent abdominal aortic constriction to induce heart failure. Four weeks after surgery, rats were then randomly divided into two groups: one group given valsartan (HF + Val) and the other group given placebo (HF + PBO). Simultaneously, the sham-operated rats were randomly given valsartan (Sham + Val) or placebo (Sham + PBO). After 4 weeks of treatment, Western blot analysis was employed to quantify CaMKII and relative calcium handling proteins (RyR2 and PLN) in all groups. Results. The deteriorated cardiac function was reversed by valsartan treatment. In ventricular muscle cells of group HF + PBO, Thr287 phosphorylation of CaMKII and S2808 phosphorylation of RyR2 and PLN were increased and S16 phosphorylation of PLN was decreased compared to the other groups, while Met281 oxidation was not significantly elevated. In addition, these changes in the expression of calcium handling proteins were ameliorated by valsartan administration. Conclusions. The phosphorylation of Thr286 is associated with the early activation of CaMKII rather than the oxidation of Met281.
Collapse
|
33
|
Hempel N, Trebak M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium 2017; 63:70-96. [PMID: 28143649 DOI: 10.1016/j.ceca.2017.01.007] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
The interplay between Ca2+ and reactive oxygen species (ROS) signaling pathways is well established, with reciprocal regulation occurring at a number of subcellular locations. Many Ca2+ channels at the cell surface and intracellular organelles, including the endoplasmic reticulum and mitochondria are regulated by redox modifications. In turn, Ca2+ signaling can influence the cellular generation of ROS, from sources such as NADPH oxidases and mitochondria. This relationship has been explored in great depth during the process of apoptosis, where surges of Ca2+ and ROS are important mediators of cell death. More recently, coordinated and localized Ca2+ and ROS transients appear to play a major role in a vast variety of pro-survival signaling pathways that may be crucial for both physiological and pathophysiological functions. While much work is required to firmly establish this Ca2+-ROS relationship in cancer, existing evidence from other disease models suggests this crosstalk is likely of significant importance in tumorigenesis. In this review, we describe the regulation of Ca2+ channels and transporters by oxidants and discuss the potential consequences of the ROS-Ca2+ interplay in tumor cells.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Pharmacology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| |
Collapse
|
34
|
Beckhauser TF, Francis-Oliveira J, De Pasquale R. Reactive Oxygen Species: Physiological and Physiopathological Effects on Synaptic Plasticity. J Exp Neurosci 2016; 10:23-48. [PMID: 27625575 PMCID: PMC5012454 DOI: 10.4137/jen.s39887] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/09/2016] [Accepted: 08/13/2016] [Indexed: 12/18/2022] Open
Abstract
In the mammalian central nervous system, reactive oxygen species (ROS) generation is counterbalanced by antioxidant defenses. When large amounts of ROS accumulate, antioxidant mechanisms become overwhelmed and oxidative cellular stress may occur. Therefore, ROS are typically characterized as toxic molecules, oxidizing membrane lipids, changing the conformation of proteins, damaging nucleic acids, and causing deficits in synaptic plasticity. High ROS concentrations are associated with a decline in cognitive functions, as observed in some neurodegenerative disorders and age-dependent decay of neuroplasticity. Nevertheless, controlled ROS production provides the optimal redox state for the activation of transductional pathways involved in synaptic changes. Since ROS may regulate neuronal activity and elicit negative effects at the same time, the distinction between beneficial and deleterious consequences is unclear. In this regard, this review assesses current research and describes the main sources of ROS in neurons, specifying their involvement in synaptic plasticity and distinguishing between physiological and pathological processes implicated.
Collapse
Affiliation(s)
- Thiago Fernando Beckhauser
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| | - José Francis-Oliveira
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| | - Roberto De Pasquale
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| |
Collapse
|
35
|
Desalegn G, Turetschek R, Kaul HP, Wienkoop S. Microbial symbionts affect Pisum sativum proteome and metabolome under Didymella pinodes infection. J Proteomics 2016; 143:173-187. [PMID: 27016040 DOI: 10.1016/j.jprot.2016.03.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/18/2016] [Accepted: 03/15/2016] [Indexed: 11/15/2022]
Abstract
UNLABELLED The long cultivation of field pea led to an enormous diversity which, however, seems to hold just little resistance against the ascochyta blight disease complex. The potential of below ground microbial symbiosis to prime the immune system of Pisum for an upcoming pathogen attack has hitherto received little attention. This study investigates the effect of beneficial microbes on the leaf proteome and metabolome as well as phenotype characteristics of plants in various symbiont interactions (mycorrhiza, rhizobia, co-inoculation, non-symbiotic) after infestation by Didymella pinodes. In healthy plants, mycorrhiza and rhizobia induced changes in RNA metabolism and protein synthesis. Furthermore, metal handling and ROS dampening was affected in all mycorrhiza treatments. The co-inoculation caused the synthesis of stress related proteins with concomitant adjustment of proteins involved in lipid biosynthesis. The plant's disease infection response included hormonal adjustment, ROS scavenging as well as synthesis of proteins related to secondary metabolism. The regulation of the TCA, amino acid and secondary metabolism including the pisatin pathway, was most pronounced in rhizobia associated plants which had the lowest infection rate and the slowest disease progression. BIOLOGICAL SIGNIFICANCE A most comprehensive study of the Pisum sativum proteome and metabolome infection response to Didymella pinodes is provided. Several distinct patterns of microbial symbioses on the plant metabolism are presented for the first time. Upon D. pinodes infection, rhizobial symbiosis revealed induced systemic resistance e.g. by an enhanced level of proteins involved in pisatin biosynthesis.
Collapse
Affiliation(s)
- G Desalegn
- University of Natural Resources and Life Sciences, Department of Crop Sciences, Austria
| | - R Turetschek
- University of Vienna, Department of Ecogenomics and Systems Biology, Austria
| | - H-P Kaul
- University of Natural Resources and Life Sciences, Department of Crop Sciences, Austria
| | - S Wienkoop
- University of Vienna, Department of Ecogenomics and Systems Biology, Austria.
| |
Collapse
|
36
|
Toussaint F, Charbel C, Allen BG, Ledoux J. Vascular CaMKII: heart and brain in your arteries. Am J Physiol Cell Physiol 2016; 311:C462-78. [PMID: 27306369 DOI: 10.1152/ajpcell.00341.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/14/2016] [Indexed: 01/02/2023]
Abstract
First characterized in neuronal tissues, the multifunctional calcium/calmodulin-dependent protein kinase II (CaMKII) is a key signaling component in several mammalian biological systems. Its unique capacity to integrate various Ca(2+) signals into different specific outcomes is a precious asset to excitable and nonexcitable cells. Numerous studies have reported roles and mechanisms involving CaMKII in brain and heart tissues. However, corresponding functions in vascular cell types (endothelium and vascular smooth muscle cells) remained largely unexplored until recently. Investigation of the intracellular Ca(2+) dynamics, their impact on vascular cell function, the regulatory processes involved and more recently the spatially restricted oscillatory Ca(2+) signals and microdomains triggered significant interest towards proteins like CaMKII. Heteromultimerization of CaMKII isoforms (four isoforms and several splice variants) expands this kinase's peculiar capacity to decipher Ca(2+) signals and initiate specific signaling processes, and thus controlling cellular functions. The physiological functions that rely on CaMKII are unsurprisingly diverse, ranging from regulating contractile state and cellular proliferation to Ca(2+) homeostasis and cellular permeability. This review will focus on emerging evidence of CaMKII as an essential component of the vascular system, with a focus on the kinase isoform/splice variants and cellular system studied.
Collapse
Affiliation(s)
- Fanny Toussaint
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Molecular and Integrative Physiology, Université de Montréal, Montreal Quebec, Canada
| | - Chimène Charbel
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology, Université de Montréal, Montreal Quebec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal Quebec, Canada; and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal Quebec, Canada
| | - Jonathan Ledoux
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal Quebec, Canada; and
| |
Collapse
|
37
|
Luo SW, Xie FX, Liu Y, Wang WN. Characterization and expression analysis of Calmodulin (CaM) in orange-spotted grouper (Epinephelus coioides) in response to Vibrio alginolyticus challenge. ECOTOXICOLOGY (LONDON, ENGLAND) 2015; 24:1775-1787. [PMID: 25956977 DOI: 10.1007/s10646-015-1467-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 06/04/2023]
Abstract
Vibrio alginolyticus containing the highly toxic extracellular product is one of the most serious threats to grouper survival and its minimum lethal dose is approximately 500 CFU/g fish body weight in grouper. To study the toxic effects of V. alginolyticus on the immune system in teleost, Calmodulin (CaM), an important molecular indicator gene, was cloned from the orange-spotted grouper (Epinephelus coioides). The full-length Ec-CaM consisted of a 5'-UTR of 103 bp, an ORF of 450 bp and a 3'-UTR of 104 bp. The Ec-CaM gene encoded a protein of 149 amino acids with an estimated molecular mass of 16.4 kDa and a predicted isoelectric point of 3.93. The deduced amino acid sequence showed that Ec-CaM contained four highly conserved EF-hand domains known to be critical for the function of CaM. Ec-CaM was widely expressed and the highest expression level was observed in liver. Following V. alginolyticus challenge, a sharp increase level of respiratory burst activity and apoptosis ratio were observed. Further analyses of CaM expression and p53 expression in liver, kidney and spleen by qRT-PCR demonstrated that the up-regulated expression of CaM and p53 were observed in the vibrio challenge group. Western blotting analysis confirmed that the Ec-CaM protein was strongly induced in liver at 12 h post-injection, while a sharp increase of p53 protein expression was observed at 24 h post-injection. These results showed CaM expression serving as a potential molecular indicator may help to assess the toxicological effects of V. alginolyticus on the ROS generation and apoptotic process in grouper.
Collapse
Affiliation(s)
- Sheng-Wei Luo
- Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Fu-Xing Xie
- Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Yuan Liu
- Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Wei-Na Wang
- Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China.
| |
Collapse
|
38
|
Abstract
SIGNIFICANCE A constant accumulation of oxidized proteins takes place during aging. Oxidation of proteins leads to a partial unfolding and, therefore, to aggregation. Protein aggregates impair the activity of cellular proteolytic systems (proteasomes, lysosomes), resulting in further accumulation of oxidized proteins. In addition, the accumulation of highly crosslinked protein aggregates leads to further oxidant formation, damage to macromolecules, and, finally, to apoptotic cell death. Furthermore, protein oxidation seems to play a role in the development of various age-related diseases, for example, neurodegenerative diseases. RECENT ADVANCES The highly oxidized lipofuscin accumulates during aging. Lipofuscin formation might cause impaired lysosomal and proteasomal degradation, metal ion accumulation, increased reactive oxygen species formation, and apoptosis. CRITICAL ISSUES It is still unclear to which extent protein oxidation is involved in the progression of aging and in the development of some age-related diseases. FUTURE DIRECTIONS An extensive knowledge of the effects of protein oxidation on the aging process and its contribution to the development of age-related diseases could enable further strategies to reduce age-related impairments. Strategies aimed at lowering aggregate formation might be a straightforward intervention to reduce age-related malfunctions of organs.
Collapse
Affiliation(s)
- Sandra Reeg
- German Institute of Human Nutrition , Nuthetal, Germany
| | - Tilman Grune
- German Institute of Human Nutrition , Nuthetal, Germany
| |
Collapse
|
39
|
Ferreira R, Wong R, Schlichter LC. KCa3.1/IK1 Channel Regulation by cGMP-Dependent Protein Kinase (PKG) via Reactive Oxygen Species and CaMKII in Microglia: An Immune Modulating Feedback System? Front Immunol 2015; 6:153. [PMID: 25904916 PMCID: PMC4389654 DOI: 10.3389/fimmu.2015.00153] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/23/2015] [Indexed: 01/09/2023] Open
Abstract
The intermediate conductance Ca2+-activated K+ channel, KCa3.1 (IK1/SK4/KCNN4) is widely expressed in the innate and adaptive immune system. KCa3.1 contributes to proliferation of activated T lymphocytes, and in CNS-resident microglia, it contributes to Ca2+ signaling, migration, and production of pro-inflammatory mediators (e.g., reactive oxygen species, ROS). KCa3.1 is under investigation as a therapeutic target for CNS disorders that involve microglial activation and T cells. However, KCa3.1 is post-translationally regulated, and this will determine when and how much it can contribute to cell functions. We previously found that KCa3.1 trafficking and gating require calmodulin (CaM) binding, and this is inhibited by cAMP kinase (PKA) acting at a single phosphorylation site. The same site is potentially phosphorylated by cGMP kinase (PKG), and in some cells, PKG can increase Ca2+, CaM activation, and ROS. Here, we addressed KCa3.1 regulation through PKG-dependent pathways in primary rat microglia and the MLS-9 microglia cell line, using perforated-patch recordings to preserve intracellular signaling. Elevating cGMP increased both the KCa3.1 current and intracellular ROS production, and both were prevented by the selective PKG inhibitor, KT5823. The cGMP/PKG-evoked increase in KCa3.1 current in intact MLS-9 microglia was mediated by ROS, mimicked by applying hydrogen peroxide (H2O2), inhibited by a ROS scavenger (MGP), and prevented by a selective CaMKII inhibitor (mAIP). Similar results were seen in alternative-activated primary rat microglia; their KCa3.1 current required PKG, ROS, and CaMKII, and they had increased ROS production that required KCa3.1 activity. The increase in current apparently did not result from direct effects on the channel open probability (Po) or Ca2+ dependence because, in inside-out patches from transfected HEK293 cells, single-channel activity was not affected by cGMP, PKG, H2O2 at normal or elevated intracellular Ca2+. The regulation pathway we have identified in intact microglia and MLS-9 cells is expected to have broad implications because KCa3.1 plays important roles in numerous cells and tissues.
Collapse
Affiliation(s)
- Roger Ferreira
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| | - Raymond Wong
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| | - Lyanne C Schlichter
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
40
|
Functional and structural changes of human erythrocyte catalase induced by cimetidine: proposed model of binding. Mol Cell Biochem 2015; 404:97-102. [DOI: 10.1007/s11010-015-2369-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/21/2015] [Indexed: 11/27/2022]
|
41
|
Drazic A, Winter J. The physiological role of reversible methionine oxidation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1367-82. [PMID: 24418392 DOI: 10.1016/j.bbapap.2014.01.001] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/17/2013] [Accepted: 01/02/2014] [Indexed: 01/04/2023]
Abstract
Sulfur-containing amino acids such as cysteine and methionine are particularly vulnerable to oxidation. Oxidation of cysteine and methionine in their free amino acid form renders them unavailable for metabolic processes while their oxidation in the protein-bound state is a common post-translational modification in all organisms and usually alters the function of the protein. In the majority of cases, oxidation causes inactivation of proteins. Yet, an increasing number of examples have been described where reversible cysteine oxidation is part of a sophisticated mechanism to control protein function based on the redox state of the protein. While for methionine the dogma is still that its oxidation inhibits protein function, reversible methionine oxidation is now being recognized as a powerful means of triggering protein activity. This mode of regulation involves oxidation of methionine to methionine sulfoxide leading to activated protein function, and inactivation is accomplished by reduction of methionine sulfoxide back to methionine catalyzed by methionine sulfoxide reductases. Given the similarity to thiol-based redox-regulation of protein function, methionine oxidation is now established as a novel mode of redox-regulation of protein function. This article is part of a Special Issue entitled: Thiol-Based Redox Processes.
Collapse
Affiliation(s)
- Adrian Drazic
- Center for Integrated Protein Science Munich (CiPS(M)) at the Department Chemie, Technische Universität München, 85747 Garching, Germany
| | - Jeannette Winter
- Center for Integrated Protein Science Munich (CiPS(M)) at the Department Chemie, Technische Universität München, 85747 Garching, Germany.
| |
Collapse
|
42
|
Teshima Y, Takahashi N, Nishio S, Saito S, Kondo H, Fukui A, Aoki K, Yufu K, Nakagawa M, Saikawa T. Production of reactive oxygen species in the diabetic heart. Roles of mitochondria and NADPH oxidase. Circ J 2013; 78:300-6. [PMID: 24334638 DOI: 10.1253/circj.cj-13-1187] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Reactive oxygen species (ROS) are the main facilitators of cardiovascular complications in diabetes mellitus (DM), and the ROS level is increased in cultured cells exposed to high glucose concentrations or in diabetic animal models. Emerging evidence shows that mitochondria and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase are dominant mechanisms of ROS production in the diabetic heart. Hyperpolarization of the mitochondrial inner membrane potentials and impaired mitochondrial function promote ROS production in the mitochondria of the diabetic heart. Uncoupling proteins are upregulated and may reduce the ROS level by depolarizing the mitochondrial inner membrane potential. NADPH oxidase is another major site of ROS production and its contribution to DM-induced ROS increase has been elucidated not only in vascular smooth muscle cells and endothelial cells, but also in cardiomyocytes. Protein kinase C, angiotensin II, and advanced glycation endproducts (AGEs)/receptor for AGEs can activate NADPH oxidase. Increased intracellular calcium level mediated via the Na(+)-H(+) exchanger and subsequent activation of Ca(2+)/calmodulin-dependent protein kinase II may also activate NADPH oxidase. This review presents the current understanding of the mechanisms of ROS production, focusing especially on the roles of mitochondria and NADPH oxidase.
Collapse
Affiliation(s)
- Yasushi Teshima
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
SIGNIFICANCE Despite recent medical advances, cardiovascular disease and heart failure (HF) continue to be major health concerns, and related mortality remains high. As a result, investigation of the mechanisms involved in the development of HF continues to be an active field of study. RECENT ADVANCES The renin-angiotensin system (RAS) and its effector molecule, angiotensin (Ang) II, affect cardiac function through both systemic and local actions, and have been shown to play a major role in cardiac remodeling and dysfunction in the failing heart. Many of the downstream effects of AngII signaling are mediated by elevated levels of reactive oxygen species (ROS) and oxidative stress, which have also been implicated in the pathology of HF. CRITICAL ISSUES Inhibitors of the RAS have proven beneficial in the treatment of patients at risk for and suffering from HF, but remain only partially effective. ROS can be generated from several different sources, and the oxidative state is normally tightly regulated in the heart. How AngII increases ROS levels and causes dysregulation of the cardiac oxidative state has been the subject of considerable interest in recent years. FUTURE DIRECTIONS A better understanding of this process and the mechanisms involved should lead to the development of more effective HF therapies and improved outcomes.
Collapse
Affiliation(s)
- Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, University of Medicine and Dentistry of New Jersey , Newark, New Jersey
| | | |
Collapse
|
44
|
Pyo CW, Choi JH, Oh SM, Choi SY. Oxidative stress-induced cyclin D1 depletion and its role in cell cycle processing. Biochim Biophys Acta Gen Subj 2013; 1830:5316-25. [PMID: 23920145 DOI: 10.1016/j.bbagen.2013.07.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 07/01/2013] [Accepted: 07/27/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cyclin D1 is immediately down-regulated in response to reactive oxygen species (ROS) and implicated in the induction of cell cycle arrest in G2 phase by an unknown mechanism. Either treatment with a protease inhibitor alone or expression of protease-resistant cyclin D1 T286A resulted in only a partial relief from the ROS-induced cell cycle arrest, indicating the presence of an additional control mechanism. METHODS Cells were exposed to hydrogen peroxide (H2O2), and analyzed to assess the changes in cyclin D1 level and its effects on cell cycle processing by kinase assay, de novo synthesis, gene silencing, and polysomal analysis, etc. RESULTS Exposure of cells to excessive H2O2 induced ubiquitin-dependent proteasomal degradation of cyclin D1, which was subsequently followed by translational repression. This dual control mechanism was found to contribute to the induction of cell cycle arrest in G2 phase under oxidative stress. Silencing of an eIF2α kinase PERK significantly retarded cyclin D1 depletion, and contributed largely to rescuing cells from G2 arrest. Also the cyclin D1 level was found to be correlated with Chk1 activity. CONCLUSIONS In addition to an immediate removal of the pre-existing cyclin D1 under oxidative stress, the following translational repression appear to be required for ensuring full depletion of cyclin D1 and cell cycle arrest. Oxidative stress-induced cyclin D1 depletion is linked to the regulation of G2/M transit via the Chk1-Cdc2 DNA damage checkpoint pathway. GENERAL SIGNIFICANCE The control of cyclin D1 is a gate keeping program to protect cells from severe oxidative damages.
Collapse
Affiliation(s)
- Chul-Woong Pyo
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | |
Collapse
|
45
|
Rokita AG, Anderson ME. New therapeutic targets in cardiology: arrhythmias and Ca2+/calmodulin-dependent kinase II (CaMKII). Circulation 2013; 126:2125-39. [PMID: 23091085 DOI: 10.1161/circulationaha.112.124990] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Adam G Rokita
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | |
Collapse
|
46
|
Bers DM, Grandi E. Human atrial fibrillation: insights from computational electrophysiological models. Trends Cardiovasc Med 2012; 21:145-50. [PMID: 22732550 DOI: 10.1016/j.tcm.2012.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 11/16/2022]
Abstract
Computational electrophysiology has proven useful to investigate the mechanisms of cardiac arrhythmias at various spatial scales, from isolated myocytes to the whole heart. This article reviews how mathematical modeling has aided our understanding of human atrial myocyte electrophysiology to study the contribution of structural and electrical remodeling to human atrial fibrillation. Potential new avenues of investigation and model development are suggested.
Collapse
Affiliation(s)
- Donald M Bers
- Department of Pharmacology, University of California at Davis, Davis, CA 95616-8636, USA.
| | | |
Collapse
|
47
|
Komiya M, Asano S, Koike N, Koga E, Igarashi J, Nakatani S, Isobe Y. Synthesis and structure based optimization of 2-(4-phenoxybenzoyl)-5-hydroxyindole as a novel CaMKII inhibitor. Bioorg Med Chem 2012; 20:6840-7. [PMID: 23088910 DOI: 10.1016/j.bmc.2012.09.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/23/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
Based on 2-(4-phenoxybenzoyl)-5-hydroxyindole (2), a novel structural class of CaMKII inhibitors were synthesized and further optimized. The strong acidity of the hydroxyl group and the lipophilic group at the 4 and 6-positions were found to be necessary for strong CaMKII inhibition. Compound 25 was identified as a promising compound with 50-fold more potent inhibitory activity for CaMKII than 2. Compound 25 also showed high selectivity for CaMKII over off-target kinases.
Collapse
Affiliation(s)
- Masafumi Komiya
- Research Division, Dainippon Sumitomo Pharma Co., Ltd, Enoki, Suita, Osaka, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Campese VM, Ye S. A vitamin-E-fortified diet reduces oxidative stress, sympathetic nerve activity, and hypertension in the phenol-renal injury model in rats. ACTA ACUST UNITED AC 2012; 1:242-50. [PMID: 20409856 DOI: 10.1016/j.jash.2007.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 04/19/2007] [Accepted: 04/20/2007] [Indexed: 02/07/2023]
Abstract
Renal injury caused by the injection of phenol in the lower pole of one kidney increases sympathetic nervous system (SNS) activity and blood pressure (BP), and these effects are mediated by increased reactive oxygen species (ROS) in brain nuclei involved in the noradrenergic control of BP. This suggests that therapy with antioxidants might be beneficial in this model. In this study, we tested the hypothesis that a vitamin (Vit)-E-enriched diet might decrease oxidative stress in the brain and result in reduced SNS activity and BP in animals with phenol-renal injury. To this end, we examined the effects of a Vit-E-fortified diet vs. a control diet on BP, norepinephrine (NE) secretion from the posterior hypothalamic nuclei (PH), and the abundance of several components of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase in the brain of rats with phenol-induced renal injury. A Vit-E-fortified diet mitigated the formation of ROS in the brain, and this was associated with reduced SNS activity and BP in rats with phenol-induced renal injury. In conclusion, antioxidants appear to be beneficial in the management of hypertension caused by renal injury and increased SNS activity.
Collapse
Affiliation(s)
- Vito M Campese
- Division of Nephrology; and the Hypertension Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
49
|
Baba H, Sueyoshi N, Shigeri Y, Ishida A, Kameshita I. Regulation of Ca2+/calmodulin-dependent protein kinase phosphatase (CaMKP) by oxidation/reduction at Cys-359. Arch Biochem Biophys 2012; 526:9-15. [DOI: 10.1016/j.abb.2012.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/12/2012] [Accepted: 06/18/2012] [Indexed: 10/28/2022]
|
50
|
Muik M, Schindl R, Fahrner M, Romanin C. Ca(2+) release-activated Ca(2+) (CRAC) current, structure, and function. Cell Mol Life Sci 2012; 69:4163-76. [PMID: 22802126 PMCID: PMC3505497 DOI: 10.1007/s00018-012-1072-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 06/07/2012] [Accepted: 06/21/2012] [Indexed: 10/28/2022]
Abstract
Store-operated Ca(2+) entry describes the phenomenon that connects a depletion of internal Ca(2+) stores to an activation of plasma membrane-located Ca(2+) selective ion channels. Tremendous progress towards the underlying molecular mechanism came with the discovery of the two respective limiting components, STIM and Orai. STIM1 represents the ER-located Ca(2+) sensor and transmits the signal of store depletion to the plasma membrane. Here it couples to and activates Orai, the highly Ca(2+)-selective pore-forming subunit of Ca(2+) release-activated Ca(2+) channels. In this review, we focus on the molecular steps that these two proteins undergo from store-depletion to their coupling, the activation, and regulation of Ca(2+) currents.
Collapse
Affiliation(s)
- Martin Muik
- Institute of Biophysics, University of Linz, Gruberstrasse 40, 4020 Linz, Austria
| | | | | | | |
Collapse
|