1
|
Guo Q, Huo Y, Liu Q, Zhou S, Xiao Y. Ruxolitinib as a CaMKII inhibitor for treatment of cardiac arrhythmias: Applications and prospects. Heart Rhythm 2025; 22:231-239. [PMID: 39111609 DOI: 10.1016/j.hrthm.2024.07.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Recent studies have highlighted the critical role of calcium/calmodulin-dependent protein kinase II (CaMKII) overactivation in the pathogenesis of various cardiac arrhythmias. Ruxolitinib, a Janus kinase inhibitor widely used for the treatment of myelofibrosis and acute graft-vs-host disease, has expanded its research horizons to include its potential as a CaMKII inhibitor in the treatment of cardiac arrhythmias. This article reviews the basic pharmacologic properties of ruxolitinib and delves into the role of CaMKII in cardiac arrhythmias, including its structural fundamentals, activation mechanisms, and association with arrhythmic conditions. Furthermore, the current state of CaMKII inhibitor research is discussed, with a special focus on the advances and clinical potential of ruxolitinib in this field. Studies indicate that ruxolitinib effectively inhibits CaMKII activity and has therapeutic potential against cardiac arrhythmias in animal models and at the cellular level. In addition, we address the critical issues that need to be resolved before the clinical application of ruxolitinib in arrhythmia treatment, including dosage concerns, long-term inhibitory effects, potential impacts on the nervous system, and efficacy across different types of arrhythmias. Future research directions involve further exploration of the clinical application potential of ruxolitinib, particularly in diseases such as heart failure, hypertrophic cardiomyopathy, dilated cardiomyopathy, and ischemic arrhythmias. In summary, the efficacy, low toxicity, and safety profile of ruxolitinib as a CaMKII inhibitor in the treatment of cardiac arrhythmias suggest a promising future for its development as a therapeutic drug in this domain.
Collapse
Affiliation(s)
- Qingbo Guo
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | - Yiran Huo
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
2
|
Wu LY, Song YJ, Zhang CL, Liu J. K V Channel-Interacting Proteins in the Neurological and Cardiovascular Systems: An Updated Review. Cells 2023; 12:1894. [PMID: 37508558 PMCID: PMC10377897 DOI: 10.3390/cells12141894] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
KV channel-interacting proteins (KChIP1-4) belong to a family of Ca2+-binding EF-hand proteins that are able to bind to the N-terminus of the KV4 channel α-subunits. KChIPs are predominantly expressed in the brain and heart, where they contribute to the maintenance of the excitability of neurons and cardiomyocytes by modulating the fast inactivating-KV4 currents. As the auxiliary subunit, KChIPs are critically involved in regulating the surface protein expression and gating properties of KV4 channels. Mechanistically, KChIP1, KChIP2, and KChIP3 promote the translocation of KV4 channels to the cell membrane, accelerate voltage-dependent activation, and slow the recovery rate of inactivation, which increases KV4 currents. By contrast, KChIP4 suppresses KV4 trafficking and eliminates the fast inactivation of KV4 currents. In the heart, IKs, ICa,L, and INa can also be regulated by KChIPs. ICa,L and INa are positively regulated by KChIP2, whereas IKs is negatively regulated by KChIP2. Interestingly, KChIP3 is also known as downstream regulatory element antagonist modulator (DREAM) because it can bind directly to the downstream regulatory element (DRE) on the promoters of target genes that are implicated in the regulation of pain, memory, endocrine, immune, and inflammatory reactions. In addition, all the KChIPs can act as transcription factors to repress the expression of genes involved in circadian regulation. Altered expression of KChIPs has been implicated in the pathogenesis of several neurological and cardiovascular diseases. For example, KChIP2 is decreased in failing hearts, while loss of KChIP2 leads to increased susceptibility to arrhythmias. KChIP3 is increased in Alzheimer's disease and amyotrophic lateral sclerosis, but decreased in epilepsy and Huntington's disease. In the present review, we summarize the progress of recent studies regarding the structural properties, physiological functions, and pathological roles of KChIPs in both health and disease. We also summarize the small-molecule compounds that regulate the function of KChIPs. This review will provide an overview and update of the regulatory mechanism of the KChIP family and the progress of targeted drug research as a reference for researchers in related fields.
Collapse
Affiliation(s)
- Le-Yi Wu
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yu-Juan Song
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Jie Liu
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen 518060, China
| |
Collapse
|
3
|
Kiessling M, Djalinac N, Voglhuber J, Ljubojevic-Holzer S. Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact. Biomedicines 2023; 11:biomedicines11030960. [PMID: 36979939 PMCID: PMC10046765 DOI: 10.3390/biomedicines11030960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
The nucleus of a cardiomyocyte has been increasingly recognized as a morphologically distinct and partially independent calcium (Ca2+) signaling microdomain, with its own Ca2+-regulatory mechanisms and important effects on cardiac gene expression. In this review, we (1) provide a comprehensive overview of the current state of research on the dynamics and regulation of nuclear Ca2+ signaling in cardiomyocytes, (2) address the role of nuclear Ca2+ in the development and progression of cardiac pathologies, such as heart failure and atrial fibrillation, and (3) discuss novel aspects of experimental methods to investigate nuclear Ca2+ handling and its downstream effects in the heart. Finally, we highlight current challenges and limitations and recommend future directions for addressing key open questions.
Collapse
Affiliation(s)
- Mara Kiessling
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
| | - Nataša Djalinac
- Department of Biology, University of Padua, 35122 Padova, Italy
| | - Julia Voglhuber
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
4
|
Siwaponanan P, Kaewkumdee P, Phromawan W, Udompunturak S, Chomanee N, Udol K, Pattanapanyasat K, Krittayaphong R. Increased expression of six-large extracellular vesicle-derived miRNAs signature for nonvalvular atrial fibrillation. J Transl Med 2022; 20:4. [PMID: 34980172 PMCID: PMC8722074 DOI: 10.1186/s12967-021-03213-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022] Open
Abstract
Backgrounds Non-valvular atrial fibrillation (AF) is the most common type of cardiac arrhythmia. AF is caused by electrophysiological abnormalities and alteration of atrial tissues, which leads to the generation of abnormal electrical impulses. Extracellular vesicles (EVs) are membrane-bound vesicles released by all cell types. Large EVs (lEVs) are secreted by the outward budding of the plasma membrane during cell activation or cell stress. lEVs are thought to act as vehicles for miRNAs to modulate cardiovascular function, and to be involved in the pathophysiology of cardiovascular diseases (CVDs), including AF. This study identified lEV-miRNAs that were differentially expressed between AF patients and non-AF controls. Methods lEVs were isolated by differential centrifugation and characterized by Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM), flow cytometry and Western blot analysis. For the discovery phase, 12 AF patients and 12 non-AF controls were enrolled to determine lEV-miRNA profile using quantitative reverse transcription polymerase chain reaction array. The candidate miRNAs were confirmed their expression in a validation cohort using droplet digital PCR (30 AF, 30 controls). Bioinformatics analysis was used to predict their target genes and functional pathways. Results TEM, NTA and flow cytometry demonstrated that lEVs presented as cup shape vesicles with a size ranging from 100 to 1000 nm. AF patients had significantly higher levels of lEVs at the size of 101–200 nm than non-AF controls. Western blot analysis was used to confirm EV markers and showed the high level of cardiomyocyte expression (Caveolin-3) in lEVs from AF patients. Nineteen miRNAs were significantly higher (> twofold, p < 0.05) in AF patients compared to non-AF controls. Six highly expressed miRNAs (miR-106b-3p, miR-590-5p, miR-339-3p, miR-378-3p, miR-328-3p, and miR-532-3p) were selected to confirm their expression. Logistic regression analysis showed that increases in the levels of these 6 highly expressed miRNAs associated with AF. The possible functional roles of these lEV-miRNAs may involve in arrhythmogenesis, cell apoptosis, cell proliferation, oxygen hemostasis, and structural remodeling in AF. Conclusion Increased expression of six lEV-miRNAs reflects the pathophysiology of AF that may provide fundamental knowledge to develop the novel biomarkers for diagnosis or monitoring the patients with the high risk of AF. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03213-6.
Collapse
Affiliation(s)
- Panjaree Siwaponanan
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pontawee Kaewkumdee
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wilasinee Phromawan
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suthipol Udompunturak
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nusara Chomanee
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kamol Udol
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rungroj Krittayaphong
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
5
|
Stress-driven cardiac calcium mishandling via a kinase-to-kinase crosstalk. Pflugers Arch 2021; 473:363-375. [PMID: 33590296 PMCID: PMC7940337 DOI: 10.1007/s00424-021-02533-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/25/2023]
Abstract
Calcium homeostasis in the cardiomyocyte is critical to the regulation of normal cardiac function. Abnormal calcium dynamics such as altered uptake by the sarcoplasmic reticulum (SR) Ca2+-ATPase and increased diastolic SR calcium leak are involved in the development of maladaptive cardiac remodeling under pathological conditions. Ca2+/calmodulin-dependent protein kinase II-δ (CaMKIIδ) is a well-recognized key molecule in calcium dysregulation in cardiomyocytes. Elevated cellular stress is known as a common feature during pathological remodeling, and c-jun N-terminal kinase (JNK) is an important stress kinase that is activated in response to intrinsic and extrinsic stress stimuli. Our lab recently identified specific actions of JNK isoform 2 (JNK2) in CaMKIIδ expression, activation, and CaMKIIδ-dependent SR Ca2+ mishandling in the stressed heart. This review focuses on the current understanding of cardiac SR calcium handling under physiological and pathological conditions as well as the newly identified contribution of the stress kinase JNK2 in CaMKIIδ-dependent SR Ca2+ abnormal mishandling. The new findings identifying dual roles of JNK2 in CaMKIIδ expression and activation are also discussed in this review.
Collapse
|
6
|
Jiang SJ, Wang W. Research progress on the role of CaMKII in heart disease. Am J Transl Res 2020; 12:7625-7639. [PMID: 33437349 PMCID: PMC7791482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
In the heart, Ca2+ participates in electrical activity and myocardial contraction, which is closely related to the generation of action potential and excitation contraction coupling (ECC) and plays an important role in various signal cascades and regulates different physiological processes. In the Ca2+ related physiological activities, CaMKII is a key downstream regulator, involving autophosphorylation and post-translational modification, and plays an important role in the excitation contraction coupling and relaxation events of cardiomyocytes. This paper reviews the relationship between CaMKII and various substances in the pathological process of myocardial apoptosis and necrosis, myocardial hypertrophy and arrhythmia, and what roles it plays in the development of disease in complex networks. This paper also introduces the drugs targeting at CaMKII to treat heart disease.
Collapse
Affiliation(s)
- Shi-Jun Jiang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Wei Wang
- Department of Cardiology, Affiliated Taihe Hospital of Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
7
|
Han JW, Kang C, Kim Y, Lee MG, Kim JY. Isoproterenol-induced hypertrophy of neonatal cardiac myocytes and H9c2 cell is dependent on TRPC3-regulated Ca V1.2 expression. Cell Calcium 2020; 92:102305. [PMID: 33069962 DOI: 10.1016/j.ceca.2020.102305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
CaV1.2 and transient receptor potential canonical channel 3 (TRPC3) are two proteins known to have important roles in pathological cardiac hypertrophy; however, such roles still remain unclear. A better understanding of these roles is important for furthering the clinical understanding of heart failure. We previously reported that Trpc3-knockout (KO) mice are resistant to pathologic hypertrophy and that their CaV1.2 protein expression is reduced. In this study, we aimed to examine the relationship between these two proteins and characterize their role in neonatal cardiomyocytes. We measured CaV1.2 expression in the hearts of wild-type (WT) and Trpc3-/- mice, and examined the effects of Trpc3 knockdown and overexpression in the rat cell line H9c2. We also compared the hypertrophic responses of neonatal cardiomyocytes cultured from Trpc3-/- mice to a representative hypertrophy-causing drug, isoproterenol (ISO), and measured the activity of nuclear factor of activated T cells 3 (NFAT3) in neonatal cardiomyocytes (NCMCs). We inhibited the L-type current with nifedipine, and measured the intracellular calcium concentration using Fura-2 with 1-oleoyl-2-acetyl-sn-glycerol (OAG)-induced Ba2+ influx. When using the Trpc3-mediated Ca2+ influx, both intracellular calcium concentration and calcium influx were reduced in Trpc3-KO myocytes. Not only was the expression of CaV1.2 greatly reduced in Trpc3-KO cardiac lysate, but the size of the CaV1.2 currents in NCMCs was also greatly reduced. When NCMCs were treated with Trpc3 siRNA, it was confirmed that the expression of CaV1.2 and the intracellular nuclear transfer activity of NFAT decreased. In H9c2 cells, the ISO activated- and verapamil inhibited- Ca2+ influxes were dramatically attenuated by Trpc3 siRNA treatment. In addition, it was confirmed that both the expression of CaV1.2 and the size of H9c2 cells were regulated according to the expression and activation level of TRPC3. We found that after stimulation with ISO, cell hypertrophy occurred in WT myocytes, while the increase in size of Trpc3-KO myocytes was greatly reduced. These results suggest that not only the cell hypertrophy process in neonatal cardiac myocytes and H9c2 cells were regulated according to the expression level of CaV1.2, but also that the expression level of CaV1.2 was regulated by TRPC3 through the activation of NFAT.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Choeun Kang
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Yonjung Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| |
Collapse
|
8
|
Cardiomyocyte calcium handling in health and disease: Insights from in vitro and in silico studies. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 157:54-75. [PMID: 32188566 DOI: 10.1016/j.pbiomolbio.2020.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/31/2019] [Accepted: 02/29/2020] [Indexed: 02/07/2023]
Abstract
Calcium (Ca2+) plays a central role in cardiomyocyte excitation-contraction coupling. To ensure an optimal electrical impulse propagation and cardiac contraction, Ca2+ levels are regulated by a variety of Ca2+-handling proteins. In turn, Ca2+ modulates numerous electrophysiological processes. Accordingly, Ca2+-handling abnormalities can promote cardiac arrhythmias via various mechanisms, including the promotion of afterdepolarizations, ion-channel modulation and structural remodeling. In the last 30 years, significant improvements have been made in the computational modeling of cardiomyocyte Ca2+ handling under physiological and pathological conditions. However, numerous questions involving the Ca2+-dependent regulation of different macromolecular complexes, cross-talk between Ca2+-dependent regulatory pathways operating over a wide range of time scales, and bidirectional interactions between electrophysiology and mechanics remain to be addressed by in vitro and in silico studies. A better understanding of disease-specific Ca2+-dependent proarrhythmic mechanisms may facilitate the development of improved therapeutic strategies. In this review, we describe the fundamental mechanisms of cardiomyocyte Ca2+ handling in health and disease, and provide an overview of currently available computational models for cardiomyocyte Ca2+ handling. Finally, we discuss important uncertainties and open questions about cardiomyocyte Ca2+ handling and highlight how synergy between in vitro and in silico studies may help to answer several of these issues.
Collapse
|
9
|
Cassambai S, Mee CJ, Renshaw D, Hussain A. Tiotropium bromide, a long acting muscarinic receptor antagonist triggers intracellular calcium signalling in the heart. Toxicol Appl Pharmacol 2019; 384:114778. [DOI: 10.1016/j.taap.2019.114778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/14/2019] [Accepted: 10/06/2019] [Indexed: 12/30/2022]
|
10
|
Abstract
Kv channel-interacting proteins (KChIPs) belong to the neuronal calcium sensor (NCS) family of Ca2+-binding EF-hand proteins. KChIPs constitute a group of specific auxiliary β-subunits for Kv4 channels, the molecular substrate of transient potassium currents in both neuronal and non-neuronal tissues. Moreover, KChIPs can interact with presenilins to control ER calcium signaling and apoptosis, and with DNA to control gene transcription. Ca2+ binding via their EF-hands, with the consequence of conformational changes, is well documented for KChIPs. Moreover, the Ca2+ dependence of the presenilin/KChIP complex may be related to Alzheimer’s disease and the Ca2+ dependence of the DNA/KChIP complex to pain sensing. However, only in few cases could the Ca2+ binding to KChIPs be directly linked to the control of excitability in nerve and muscle cells known to express Kv4/KChIP channel complexes. This review summarizes current knowledge about the Ca2+ binding properties of KChIPs and the Ca2+ dependencies of macromolecular complexes containing KChIPs, including those with presenilins, DNA and especially Kv4 channels. The respective physiological or pathophysiolgical roles of Ca2+ binding to KChIPs are discussed.
Collapse
Affiliation(s)
- Robert Bähring
- a Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin , Universitätsklinikum Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
11
|
Li MCH, O'Brien TJ, Todaro M, Powell KL. Acquired cardiac channelopathies in epilepsy: Evidence, mechanisms, and clinical significance. Epilepsia 2019; 60:1753-1767. [PMID: 31353444 DOI: 10.1111/epi.16301] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 12/13/2022]
Abstract
There is growing evidence that cardiac dysfunction in patients with chronic epilepsy could play a pathogenic role in sudden unexpected death in epilepsy (SUDEP). Recent animal studies have revealed that epilepsy secondarily alters the expression of cardiac ion channels alongside abnormal cardiac electrophysiology and remodeling. These molecular findings represent novel evidence for an acquired cardiac channelopathy in epilepsy, distinct from inherited ion channels mutations associated with cardiocerebral phenotypes. Specifically, seizure activity has been shown to alter the messenger RNA (mRNA) and protein expression of voltage-gated sodium channels (Nav 1.1, Nav 1.5), voltage-gated potassium channels (Kv 4.2, Kv 4.3), sodium-calcium exchangers (NCX1), and nonspecific cation-conducting channels (HCN2, HCN4). The pathophysiology may involve autonomic dysfunction and structural cardiac disease, as both are independently associated with epilepsy and ion channel dysregulation. Indeed, in vivo and in vitro studies of cardiac pathology reveal a complex network of signaling pathways and transcription factors regulating ion channel expression in the setting of sympathetic overactivity, cardiac failure, and hypertrophy. Other mechanisms such as circulating inflammatory mediators or exogenous effects of antiepileptic medications lack evidence. Moreover, an acquired cardiac channelopathy may underlie the electrophysiologic cardiac abnormalities seen in chronic epilepsy, potentially contributing to the increased risk of malignant arrhythmias and sudden death. Therefore, further investigation is necessary to establish whether cardiac ion channel dysregulation similarly occurs in patients with epilepsy, and to characterize any pathogenic relationship with SUDEP.
Collapse
Affiliation(s)
- Michael C H Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Marian Todaro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Kim L Powell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Néant I, Haiech J, Kilhoffer MC, Aulestia FJ, Moreau M, Leclerc C. Ca 2+-Dependent Transcriptional Repressors KCNIP and Regulation of Prognosis Genes in Glioblastoma. Front Mol Neurosci 2018; 11:472. [PMID: 30618619 PMCID: PMC6305344 DOI: 10.3389/fnmol.2018.00472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/04/2018] [Indexed: 12/18/2022] Open
Abstract
Glioblastomas (GBMs) are the most aggressive and lethal primary astrocytic tumors in adults, with very poor prognosis. Recurrence in GBM is attributed to glioblastoma stem-like cells (GSLCs). The behavior of the tumor, including proliferation, progression, invasion, and significant resistance to therapies, is a consequence of the self-renewing properties of the GSLCs, and their high resistance to chemotherapies have been attributed to their capacity to enter quiescence. Thus, targeting GSLCs may constitute one of the possible therapeutic challenges to significantly improve anti-cancer treatment regimens for GBM. Ca2+ signaling is an important regulator of tumorigenesis in GBM, and the transition from proliferation to quiescence involves the modification of the kinetics of Ca2+ influx through store-operated channels due to an increased capacity of the mitochondria of quiescent GSLC to capture Ca2+. Therefore, the identification of new therapeutic targets requires the analysis of the calcium-regulated elements at transcriptional levels. In this review, we focus onto the direct regulation of gene expression by KCNIP proteins (KCNIP1–4). These proteins constitute the class E of Ca2+ sensor family with four EF-hand Ca2+-binding motifs and control gene transcription directly by binding, via a Ca2+-dependent mechanism, to specific DNA sites on target genes, called downstream regulatory element (DRE). The presence of putative DRE sites on genes associated with unfavorable outcome for GBM patients suggests that KCNIP proteins may contribute to the alteration of the expression of these prognosis genes. Indeed, in GBM, KCNIP2 expression appears to be significantly linked to the overall survival of patients. In this review, we summarize the current knowledge regarding the quiescent GSLCs with respect to Ca2+ signaling and discuss how Ca2+via KCNIP proteins may affect prognosis genes expression in GBM. This original mechanism may constitute the basis of the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Isabelle Néant
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Jacques Haiech
- Laboratoire d'Excellence Medalis, CNRS, LIT UMR 7200, Université de Strasbourg, Strasbourg, France
| | - Marie-Claude Kilhoffer
- Laboratoire d'Excellence Medalis, CNRS, LIT UMR 7200, Université de Strasbourg, Strasbourg, France
| | - Francisco J Aulestia
- Department of Basic Science and Craniofacial Biology, NYU College of Dentistry, New York, NY, United States
| | - Marc Moreau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Catherine Leclerc
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| |
Collapse
|
13
|
Calcium and Ca 2+/Calmodulin-dependent kinase II as targets for helminth parasite control. Biochem Soc Trans 2018; 46:1743-1751. [PMID: 30420417 DOI: 10.1042/bst20180480] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/07/2018] [Accepted: 10/01/2018] [Indexed: 11/17/2022]
Abstract
In eukaryotes, effective calcium homeostasis is critical for many key biological processes. There is an added level of complexity in parasites, particularly multicellular helminth worms, which modulate calcium levels while inhabiting the host microenvironment. Parasites ensure efficient calcium homeostasis through gene products, such as the calmodulin-dependent kinases (CaMK), the main focus of this review. The importance of CaMK is becoming increasingly apparent from recent functional studies of helminth and protozoan parasites. Investigations on the molecular regulation of calcium and the role of CaMK are important for both supplementing current drug regimens and finding new antiparasitic compounds. Whereas calcium regulators, including CaMK, are well characterised in mammalian systems, knowledge of their functional properties in parasites is increasing but is still in its infancy.
Collapse
|
14
|
Kinnunen SM, Tölli M, Välimäki MJ, Gao E, Szabo Z, Rysä J, Ferreira MPA, Ohukainen P, Serpi R, Correia A, Mäkilä E, Salonen J, Hirvonen J, Santos HA, Ruskoaho H. Cardiac Actions of a Small Molecule Inhibitor Targeting GATA4-NKX2-5 Interaction. Sci Rep 2018; 8:4611. [PMID: 29545582 PMCID: PMC5854571 DOI: 10.1038/s41598-018-22830-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023] Open
Abstract
Transcription factors are fundamental regulators of gene transcription, and many diseases, such as heart diseases, are associated with deregulation of transcriptional networks. In the adult heart, zinc-finger transcription factor GATA4 is a critical regulator of cardiac repair and remodelling. Previous studies also suggest that NKX2-5 plays function role as a cofactor of GATA4. We have recently reported the identification of small molecules that either inhibit or enhance the GATA4–NKX2-5 transcriptional synergy. Here, we examined the cardiac actions of a potent inhibitor (3i-1000) of GATA4–NKX2-5 interaction in experimental models of myocardial ischemic injury and pressure overload. In mice after myocardial infarction, 3i-1000 significantly improved left ventricular ejection fraction and fractional shortening, and attenuated myocardial structural changes. The compound also improved cardiac function in an experimental model of angiotensin II -mediated hypertension in rats. Furthermore, the up-regulation of cardiac gene expression induced by myocardial infarction and ischemia reduced with treatment of 3i-1000 or when micro- and nanoparticles loaded with 3i-1000 were injected intramyocardially or intravenously, respectively. The compound inhibited stretch- and phenylephrine-induced hypertrophic response in neonatal rat cardiomyocytes. These results indicate significant potential for small molecules targeting GATA4–NKX2-5 interaction to promote myocardial repair after myocardial infarction and other cardiac injuries.
Collapse
Affiliation(s)
- Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Marja Tölli
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Erhe Gao
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States of America
| | - Zoltan Szabo
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mónica P A Ferreira
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Raisa Serpi
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland. .,Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
15
|
Holmberg J, Bhattachariya A, Alajbegovic A, Rippe C, Ekman M, Dahan D, Hien TT, Boettger T, Braun T, Swärd K, Hellstrand P, Albinsson S. Loss of Vascular Myogenic Tone in miR-143/145 Knockout Mice Is Associated With Hypertension-Induced Vascular Lesions in Small Mesenteric Arteries. Arterioscler Thromb Vasc Biol 2018; 38:414-424. [PMID: 29217510 DOI: 10.1161/atvbaha.117.310499] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Pressure-induced myogenic tone is involved in autoregulation of local blood flow and confers protection against excessive pressure levels in small arteries and capillaries. Myogenic tone is dependent on smooth muscle microRNAs (miRNAs), but the identity of these miRNAs is unclear. Furthermore, the consequences of altered myogenic tone for hypertension-induced damage to small arteries are not well understood. APPROACH AND RESULTS The importance of smooth muscle-enriched microRNAs, miR-143/145, for myogenic tone was evaluated in miR-143/145 knockout mice. Furthermore, hypertension-induced vascular injury was evaluated in mesenteric arteries in vivo after angiotensin II infusion. Myogenic tone was abolished in miR-143/145 knockout mesenteric arteries, whereas contraction in response to calyculin A and potassium chloride was reduced by ≈30%. Furthermore, myogenic responsiveness was potentiated by angiotensin II in wild-type but not in knockout mice. Angiotensin II administration in vivo elevated systemic blood pressure in both genotypes. Hypertensive knockout mice developed severe vascular lesions characterized by vascular inflammation, adventitial fibrosis, and neointimal hyperplasia in small mesenteric arteries. This was associated with depolymerization of actin filaments and fragmentation of the elastic laminae at the sites of vascular lesions. CONCLUSIONS This study demonstrates that miR-143/145 expression is essential for myogenic responsiveness. During hypertension, loss of myogenic tone results in potentially damaging levels of mechanical stress and detrimental effects on small arteries. The results presented herein provide novel insights into the pathogenesis of vascular disease and emphasize the importance of controlling mechanical factors to maintain structural integrity of the vascular wall.
Collapse
Affiliation(s)
- Johan Holmberg
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Anirban Bhattachariya
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Azra Alajbegovic
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Catarina Rippe
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Mari Ekman
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Diana Dahan
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Tran Thi Hien
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Thomas Boettger
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Thomas Braun
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Karl Swärd
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Per Hellstrand
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun)
| | - Sebastian Albinsson
- From the Department of Experimental Medical Science, Lund University, Sweden (J.H., A.B., A.A., C.R., M.E., D.D., T.T.H., K.S., P.H., S.A.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T. Boettger, T. Braun).
| |
Collapse
|
16
|
Dewenter M, von der Lieth A, Katus HA, Backs J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ Res 2017; 121:1000-1020. [DOI: 10.1161/circresaha.117.310355] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium (Ca
2+
) is a universal regulator of various cellular functions. In cardiomyocytes, Ca
2+
is the central element of excitation–contraction coupling, but also impacts diverse signaling cascades and influences the regulation of gene expression, referred to as excitation–transcription coupling. Disturbances in cellular Ca
2+
-handling and alterations in Ca
2+
-dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with several excitation–transcription coupling pathways shown to be critically involved in structural and functional remodeling processes. Thus, targeting Ca
2+
-dependent transcriptional pathways might offer broad therapeutic potential. In this article, we (1) review cytosolic and nuclear Ca
2+
dynamics in cardiomyocytes with respect to their impact on Ca
2+
-dependent signaling, (2) give an overview on Ca
2+
-dependent transcriptional pathways in cardiomyocytes, and (3) discuss implications of excitation–transcription coupling in the diseased heart.
Collapse
Affiliation(s)
- Matthias Dewenter
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Albert von der Lieth
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Hugo A. Katus
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Johannes Backs
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| |
Collapse
|
17
|
Mutikainen M, Tuomainen T, Naumenko N, Huusko J, Smirin B, Laidinen S, Kokki K, Hynynen H, Ylä-Herttuala S, Heinäniemi M, Ruas JL, Tavi P. Peroxisome proliferator-activated receptor-γ coactivator 1 α1 induces a cardiac excitation-contraction coupling phenotype without metabolic remodelling. J Physiol 2017; 594:7049-7071. [PMID: 27716916 DOI: 10.1113/jp272847] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Transcriptional co-activator PGC-1α1 has been shown to regulate energy metabolism and to mediate metabolic adaptations in pathological and physiological cardiac hypertrophy but other functional implications of PGC-1α1 expression are not known. Transgenic PGC-1α1 overexpression within the physiological range in mouse heart induces purposive changes in contractile properties, electrophysiology and calcium signalling but does not induce substantial metabolic remodelling. The phenotype of the PGC-1α1 transgenic mouse heart recapitulates most of the functional modifications usually associated with the exercise-induced heart phenotype, but does not protect the heart against load-induced pathological hypertrophy. Transcriptional effects of PGC-1α1 show clear dose-dependence with diverse changes in genes in circadian clock, heat shock, excitability, calcium signalling and contraction pathways at low overexpression levels, while metabolic genes are recruited at much higher PGC-1α1 expression levels. These results imply that the physiological role of PGC-1α1 is to promote a beneficial excitation-contraction coupling phenotype in the heart. ABSTRACT The transcriptional coactivator PGC-1α1 has been identified as a central factor mediating metabolic adaptations of the heart. However, to what extent physiological changes in PGC-1α1 expression levels actually contribute to the functional adaptation of the heart is still mostly unresolved. The aim of this study was to characterize the transcriptional and functional effects of physiologically relevant, moderate PGC-1α1 expression in the heart. In vivo and ex vivo physiological analysis shows that expression of PGC-1α1 within a physiological range in mouse heart does not induce the expected metabolic alterations, but instead induces a unique excitation-contraction (EC) coupling phenotype recapitulating features typically seen in physiological hypertrophy. Transcriptional screening of PGC-1α1 overexpressing mouse heart and myocyte cultures with higher, acute adenovirus-induced PGC-1α1 expression, highlights PGC-1α1 as a transcriptional coactivator with a number of binding partners in various pathways (such as heat shock factors and the circadian clock) through which it acts as a pleiotropic transcriptional regulator in the heart, to both augment and repress the expression of its target genes in a dose-dependent fashion. At low levels of overexpression PGC-1α1 elicits a diverse transcriptional response altering the expression state of circadian clock, heat shock, excitability, calcium signalling and contraction pathways, while metabolic targets of PGC-1α1 are recruited at higher PGC-1α1 expression levels. Together these findings demonstrate that PGC-1α1 elicits a dual effect on cardiac transcription and phenotype. Further, our results imply that the physiological role of PGC-1α1 is to promote a beneficial EC coupling phenotype in the heart.
Collapse
Affiliation(s)
- Maija Mutikainen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tomi Tuomainen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nikolay Naumenko
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Boris Smirin
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Svetlana Laidinen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Krista Kokki
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Heidi Hynynen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
18
|
Groen C, Bähring R. Modulation of human Kv4.3/KChIP2 channel inactivation kinetics by cytoplasmic Ca 2. Pflugers Arch 2017; 469:1457-1470. [PMID: 28735419 DOI: 10.1007/s00424-017-2039-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
Abstract
The transient outward current (I to) in the human heart is mediated by Kv4.3 channels complexed with Kv channel interacting protein (KChIP) 2, a cytoplasmic Ca2+-binding EF-hand protein known to modulate Kv4.3 inactivation gating upon heterologous co-expression. We studied Kv4.3 channels co-expressed with wild-type (wt) or EF-hand-mutated (ΔEF) KChIP2 in human embryonic kidney (HEK) 293 cells. Co-expression took place in the absence or presence of BAPTA-AM, and macroscopic currents were recorded in the whole-cell patch-clamp configuration with different free Ca2+ concentrations in the patch-pipette. Our data indicate that Ca2+ is not necessary for Kv4.3/KChIP2 complex formation. The Kv4.3/KChIP2-mediated current decay was faster and the recovery of Kv4.3/KChIP2 channels from inactivation slower with 50 μM Ca2+ than with BAPTA (nominal Ca2+-free) in the patch-pipette. The apparent Ca2+-mediated slowing of recovery kinetics was still observed when EF-hand 4 of KChIP2 was mutated (ΔEF4) but not when EF-hand 2 (ΔEF2) was mutated, and turned into a Ca2+-mediated acceleration of recovery kinetics when EF-hand 3 (ΔEF3) was mutated. In the presence of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN-93 cytoplasmic Ca2+ (50 μM) induced an acceleration of Kv4.3/KChIP2 recovery kinetics, which was still observed when EF-hand 2 was mutated (ΔEF2) but not when EF-hand 3 (ΔEF3) or EF-hand 4 (ΔEF4) was mutated. Our results support the notion that binding of Ca2+ to KChIP2 EF-hands can acutely modulate Kv4.3/KChIP2 channel inactivation gating, but the Ca2+-dependent gating modulation depends on CaMKII action. Our findings speak for an acute modulation of I to kinetics and frequency-dependent I to availability in cardiomyocytes under conditions with elevated Ca2+ levels and CaMKII activity.
Collapse
Affiliation(s)
- Christiane Groen
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Robert Bähring
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
19
|
Nassal DM, Wan X, Liu H, Maleski D, Ramirez-Navarro A, Moravec CS, Ficker E, Laurita KR, Deschênes I. KChIP2 is a core transcriptional regulator of cardiac excitability. eLife 2017; 6. [PMID: 28263709 PMCID: PMC5338919 DOI: 10.7554/elife.17304] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 02/19/2017] [Indexed: 11/18/2022] Open
Abstract
Arrhythmogenesis from aberrant electrical remodeling is a primary cause of death among patients with heart disease. Amongst a multitude of remodeling events, reduced expression of the ion channel subunit KChIP2 is consistently observed in numerous cardiac pathologies. However, it remains unknown if KChIP2 loss is merely a symptom or involved in disease development. Using rat and human derived cardiomyocytes, we identify a previously unobserved transcriptional capacity for cardiac KChIP2 critical in maintaining electrical stability. Through interaction with genetic elements, KChIP2 transcriptionally repressed the miRNAs miR-34b and miR-34c, which subsequently targeted key depolarizing (INa) and repolarizing (Ito) currents altered in cardiac disease. Genetically maintaining KChIP2 expression or inhibiting miR-34 under pathologic conditions restored channel function and moreover, prevented the incidence of reentrant arrhythmias. This identifies the KChIP2/miR-34 axis as a central regulator in developing electrical dysfunction and reveals miR-34 as a therapeutic target for treating arrhythmogenesis in heart disease. DOI:http://dx.doi.org/10.7554/eLife.17304.001 The heart pumps blood throughout the body to provide oxygen and nourishment. To do so, proteins in the heart create electrical signals that tell the heart muscles to contract in a coordinated manner. Heart disease can cause cells to lose control of the production or activity of these proteins, creating disorganized electrical signals called arrhythmias that interfere with the heart’s ability to pump. Sometimes these arrhythmias lead to sudden death. Researchers do not know exactly what triggers these changes in the heart’s normal electrical rhythms. This has made it difficult to develop strategies to prevent these disruptions or to fix them when they occur. By studying rat and human heart cells, Nassal et al. now show that a protein called KChIP2 stops working properly during heart disease. Most importantly, because of the decreased level of KChIP2 in heart disease, KChIP2 loses the ability to restrict the production of two microRNA molecules – a role that KChIP2 was not previously known to perform. This loss of activity sets off a cascade of signals that worsens the balance of electrical activity in the heart cells, creating arrhythmias. Treatments that restored proper levels of the fully working KChIP2 protein to the heart cells or that blocked the signals set off by a lack of KChIP2 returned the electrical activity of the cells back to normal. This also stopped the development of arrhythmias. Further studies are now needed to investigate whether these treatments have the same effects in living mammals. If effective, this could ultimately lead to new treatments for heart diseases and arrhythmias. DOI:http://dx.doi.org/10.7554/eLife.17304.002
Collapse
Affiliation(s)
- Drew M Nassal
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, United States
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Haiyan Liu
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Danielle Maleski
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Angelina Ramirez-Navarro
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Christine S Moravec
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, United States
| | - Eckhard Ficker
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Kenneth R Laurita
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Isabelle Deschênes
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, United States
| |
Collapse
|
20
|
Wongrakpanich A, Morris AS, Geary SM, Joiner MLA, Salem AK. Surface-modified particles loaded with CaMKII inhibitor protect cardiac cells against mitochondrial injury. Int J Pharm 2017; 520:275-283. [PMID: 28167264 DOI: 10.1016/j.ijpharm.2017.01.061] [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] [Received: 10/07/2016] [Revised: 01/16/2017] [Accepted: 01/28/2017] [Indexed: 12/30/2022]
Abstract
An excess of calcium (Ca2+) influx into mitochondria during mitochondrial re-energization is one of the causes of myocardial cell death during ischemic/reperfusion injury. This overload of Ca2+ triggers the mitochondrial permeability transition pore (mPTP) opening which leads to programmed cell death. During the ischemic/reperfusion stage, the activated Ca2+/calmodulin-dependent protein kinase II (CaMKII) enzyme is responsible for Ca2+ influx. To reduce CaMKII-related cell death, sub-micron particles composed of poly(lactic-co-glycolic acid) (PLGA), loaded with a CaMKII inhibitor peptide were fabricated. The CaMKII inhibitor peptide-loaded (CIP) particles were coated with a mitochondria targeting moiety, triphenylphosphonium cation (TPP), which allowed the particles to accumulate and release the peptide inside mitochondria to inhibit CaMKII activity. The fluorescently labeled TPP-CIP was taken up by mitochondria and successfully reduced reactive oxygen species (ROS) caused by Isoprenaline (ISO) in a differentiated rat cardiomyocyte-like cell line. When cells were treated with TPP-CIP prior to ISO exposure, they maintained mitochondrial membrane potential. The TPP-CIP protected cells from ISO-induced ROS production and decreased mitochondrial membrane potential. Thus, TPP-CIP has the potential to be used in protection against ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Amaraporn Wongrakpanich
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Angie S Morris
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Mei-Ling A Joiner
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, United States.
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
21
|
Alajbegovic A, Turczyńska KM, Hien TT, Cidad P, Swärd K, Hellstrand P, Della Corte A, Forte A, Albinsson S. Regulation of microRNA expression in vascular smooth muscle by MRTF-A and actin polymerization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:1088-1098. [PMID: 27939432 DOI: 10.1016/j.bbamcr.2016.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 12/11/2022]
Abstract
The dynamic properties of the actin cytoskeleton in smooth muscle cells play an important role in a number of cardiovascular disease states. The state of actin does not only mediate mechanical stability and contractile function but can also regulate gene expression via myocardin related transcription factors (MRTFs). These transcriptional co-activators regulate genes encoding contractile and cytoskeletal proteins in smooth muscle. Regulation of small non-coding microRNAs (miRNAs) by actin polymerization may mediate some of these effects. MiRNAs are short non-coding RNAs that modulate gene expression by post-transcriptional regulation of target messenger RNA. In this study we aimed to determine a profile of miRNAs that were 1) regulated by actin/MRTF-A, 2) associated with the contractile smooth muscle phenotype and 3) enriched in muscle cells. This analysis was performed using cardiovascular disease-focused miRNA arrays in both mouse and human cells. The potential clinical importance of actin polymerization in aortic aneurysm was evaluated using biopsies from mildly dilated human thoracic aorta in patients with stenotic tricuspid or bicuspid aortic valve. By integrating information from multiple qPCR based miRNA arrays we identified a group of five miRNAs (miR-1, miR-22, miR-143, miR-145 and miR-378a) that were sensitive to actin polymerization and MRTF-A overexpression in both mouse and human vascular smooth muscle. With the exception of miR-22, these miRNAs were also relatively enriched in striated and/or smooth muscle containing tissues. Actin polymerization was found to be dramatically reduced in the aorta from patients with mild aortic dilations. This was associated with a decrease in actin/MRTF-regulated miRNAs. In conclusion, the transcriptional co-activator MRTF-A and actin polymerization regulated a subset of miRNAs in vascular smooth muscle. Identification of novel miRNAs regulated by actin/MRTF-A may provide further insight into the mechanisms underlying vascular disease states, such as aortic aneurysm, as well as novel ideas regarding therapeutic strategies. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Azra Alajbegovic
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Tran Thi Hien
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Amalia Forte
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | | |
Collapse
|
22
|
Ji Y, Guo X, Zhang Z, Huang Z, Zhu J, Chen QH, Gui L. CaMKIIδ meditates phenylephrine induced cardiomyocyte hypertrophy through store-operated Ca 2+ entry. Cardiovasc Pathol 2016; 27:9-17. [PMID: 27940402 DOI: 10.1016/j.carpath.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/10/2016] [Accepted: 11/18/2016] [Indexed: 01/01/2023] Open
Abstract
Evidence suggests that store-operated Ca2+ entry (SOCE) is involved in the hypertrophy of cardiomyocytes. The signaling mechanisms of SOCE contributing to cardiac hypertrophy following phenylephrine (PE) stimulation are not fully understood. Ca2+/calmodulin-dependent protein kinase II δ (CaMKIIδ) plays an important role in regulating intracellular Ca2+ hemostasis and function in the cardimyocytes. This study is aimed to determine the role of CaMKIIδ in regulating the PE-induced myocardial hypertrophy and the associated molecular signaling mechanisms. We used primary cultures of neonatal cardimyocytes isolated from the left ventricle of Sprague Dawley rats to investigate the effects of CaMKIIδ on myocardial hypertrophy and intracellular Ca2+ mobilization. We found that the expression of CaMKIIδ was enhanced in PE-induced hypertrophic cardiomyocytes. CaMKIIδ siRNA, CaMKII inhibitor KN93, and SOCE blocker BTP2 attenuated the increase in the expression of CaMKIIδ and normalized the hypertrophic markers, atrial natriuretic peptide and brain natriuretic peptide, and size of cardiomyocytes induced by PE stimulation. The protein level of stromal interaction molecule 1 and Orai1, the essential components of the SOCE, is also enhanced in hypertrophic cardiomyocytes, which were normalized by CaMKIIδ siRNA and KN93 treatment. Hypertrophic cardiomyocytes showed an increase in the peak of Ca2+ transient following store depletion, which was inhibited by SOCE blocker BTP2, CaMKIIδ siRNA, and KN93. The Ca2+ currents through Ca2+ release-activated Ca2+ channels were increased in PE-treated cardiomyocytes and were attenuated by CaMKIIδ siRNA and KN93. These data indicate that PE-induced myocardial hypertrophy requires a complex signaling pathway that involves activation of both CaMKIIδ and SOCE. In conclusion, these studies reveal that up-regulation of CaMKIIδ may contribute to the PE-induced myocardial hypertrophy through the activation of SOCE expressed in the cardiomyocytes.
Collapse
Affiliation(s)
- Yawei Ji
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Xin Guo
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Zhe Zhang
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Zhuyun Huang
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Jianghua Zhu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA
| | - Le Gui
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China.
| |
Collapse
|
23
|
Potassium Channel Interacting Protein 2 (KChIP2) is not a transcriptional regulator of cardiac electrical remodeling. Sci Rep 2016; 6:28760. [PMID: 27349185 PMCID: PMC4923891 DOI: 10.1038/srep28760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022] Open
Abstract
The heart-failure relevant Potassium Channel Interacting Protein 2 (KChIP2) augments CaV1.2 and KV4.3. KChIP3 represses CaV1.2 transcription in cardiomyocytes via interaction with regulatory DNA elements. Hence, we tested nuclear presence of KChIP2 and if KChIP2 translocates into the nucleus in a Ca2+ dependent manner. Cardiac biopsies from human heart-failure patients and healthy donor controls showed that nuclear KChIP2 abundance was significantly increased in heart failure; however, this was secondary to a large variation of total KChIP2 content. Administration of ouabain did not increase KChIP2 content in nuclear protein fractions in anesthetized mice. KChIP2 was expressed in cell lines, and Ca2+ ionophores were applied in a concentration- and time-dependent manner. The cell lines had KChIP2-immunoreactive protein in the nucleus in the absence of treatments to modulate intracellular Ca2+ concentration. Neither increasing nor decreasing intracellular Ca2+ concentrations caused translocation of KChIP2. Microarray analysis did not identify relief of transcriptional repression in murine KChIP2−/− heart samples. We conclude that although there is a baseline presence of KChIP2 in the nucleus both in vivo and in vitro, KChIP2 does not directly regulate transcriptional activity. Moreover, the nuclear transport of KChIP2 is not dependent on Ca2+. Thus, KChIP2 does not function as a conventional transcription factor in the heart.
Collapse
|
24
|
Kalyanavenkataraman S, Nanjan P, Banerji A, Nair BG, Kumar GB. Discovery of arjunolic acid as a novel non-zinc binding carbonic anhydrase II inhibitor. Bioorg Chem 2016; 66:72-9. [PMID: 27038848 DOI: 10.1016/j.bioorg.2016.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/04/2016] [Accepted: 03/23/2016] [Indexed: 11/29/2022]
Abstract
Elevated levels of carbonic anhydrase II (CA II) have been shown to be associated with cardiac hypertrophy and heart failure. Although arjunolic acid (AA) has a diverse range of therapeutic applications including cardio-protection, there have been no reports on the effect of AA on CA II. The present study describes for the first time, the novel zinc independent inhibition of CA II by AA. The molecular docking studies of AA indicated that the hydroxyl group at C2 of the A-ring, which hydrogen bonds with the catalytic site residues (His64, Asn62 and Asn67), along with the gem-dimethyl group at C20 of the E-ring, greatly influences the inhibitory activity, independent of the catalytic zinc, unlike the inhibition observed with most CA II inhibitors. Among the triterpenoids tested viz. arjunolic acid, arjunic acid, asiatic acid, oleanolic acid and ursolic acid, AA was the most potent in inhibiting CA II in vitro with an IC50 of 9μM. It was interesting to note, that in spite of exhibiting very little differences in their structures, these triterpenoids exhibited vast differences in their inhibitory activities, with IC50 values ranging from 9μM to as high as 333μM. Furthermore, AA also inhibited the cytosolic activity of CA in H9c2 cardiomyocytes, as reflected by the decrease in acidification of the intracellular pH (pHi). The decreased acidification reduced the intracellular calcium levels, which further prevented the mitochondrial membrane depolarization. Thus, these studies provide a better understanding for establishing the novel molecular mechanism involved in CA II inhibition by the non-zinc binding inhibitor AA.
Collapse
Affiliation(s)
| | - Pandurangan Nanjan
- Amrita School of Biotechnology, Amrita University, Amritapuri Campus, Clappana P.O., Kollam 690 525, Kerala, India
| | - Asoke Banerji
- Amrita School of Biotechnology, Amrita University, Amritapuri Campus, Clappana P.O., Kollam 690 525, Kerala, India
| | - Bipin G Nair
- Amrita School of Biotechnology, Amrita University, Amritapuri Campus, Clappana P.O., Kollam 690 525, Kerala, India
| | - Geetha B Kumar
- Amrita School of Biotechnology, Amrita University, Amritapuri Campus, Clappana P.O., Kollam 690 525, Kerala, India.
| |
Collapse
|
25
|
Mellström B, Kastanauskaite A, Knafo S, Gonzalez P, Dopazo XM, Ruiz-Nuño A, Jefferys JGR, Zhuo M, Bliss TVP, Naranjo JR, DeFelipe J. Specific cytoarchitectureal changes in hippocampal subareas in daDREAM mice. Mol Brain 2016; 9:22. [PMID: 26928278 PMCID: PMC4772309 DOI: 10.1186/s13041-016-0204-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/22/2016] [Indexed: 11/20/2022] Open
Abstract
Background Transcriptional repressor DREAM (downstream regulatory element antagonist modulator) is a Ca2+-binding protein that regulates Ca2+ homeostasis through gene regulation and protein-protein interactions. It has been shown that a dominant active form (daDREAM) is implicated in learning-related synaptic plasticity such as LTP and LTD in the hippocampus. Neuronal spines are reported to play important roles in plasticity and memory. However, the possible role of DREAM in spine plasticity has not been reported. Results Here we show that potentiating DREAM activity, by overexpressing daDREAM, reduced dendritic basal arborization and spine density in CA1 pyramidal neurons and increased spine density in dendrites in dentate gyrus granule cells. These microanatomical changes are accompanied by significant modifications in the expression of specific genes encoding the cytoskeletal proteins Arc, Formin 1 and Gelsolin in daDREAM hippocampus. Conclusions Our results strongly suggest that DREAM plays an important role in structural plasticity in the hippocampus. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0204-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Britt Mellström
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Asta Kastanauskaite
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Biomedical Technology Center, Politecnica University Madrid, Madrid, Spain.
| | - Shira Knafo
- Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Present address: IkerBasque Basque Foundation for Science and BioCruces, Health Research Institute, Bizkaia, Spain.
| | - Paz Gonzalez
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Xose M Dopazo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Ana Ruiz-Nuño
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | - John G R Jefferys
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | - Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada. .,Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Tim V P Bliss
- MRC National Institutes for Medical Research, Mill Hill, London, UK.
| | - Jose R Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Javier DeFelipe
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Biomedical Technology Center, Politecnica University Madrid, Madrid, Spain.
| |
Collapse
|
26
|
Hien TT, Turczyńska KM, Dahan D, Ekman M, Grossi M, Sjögren J, Nilsson J, Braun T, Boettger T, Garcia-Vaz E, Stenkula K, Swärd K, Gomez MF, Albinsson S. Elevated Glucose Levels Promote Contractile and Cytoskeletal Gene Expression in Vascular Smooth Muscle via Rho/Protein Kinase C and Actin Polymerization. J Biol Chem 2016; 291:3552-68. [PMID: 26683376 PMCID: PMC4751395 DOI: 10.1074/jbc.m115.654384] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 12/17/2015] [Indexed: 12/22/2022] Open
Abstract
Both type 1 and type 2 diabetes are associated with increased risk of cardiovascular disease. This is in part attributed to the effects of hyperglycemia on vascular endothelial and smooth muscle cells, but the underlying mechanisms are not fully understood. In diabetic animal models, hyperglycemia results in hypercontractility of vascular smooth muscle possibly due to increased activation of Rho-kinase. The aim of the present study was to investigate the regulation of contractile smooth muscle markers by glucose and to determine the signaling pathways that are activated by hyperglycemia in smooth muscle cells. Microarray, quantitative PCR, and Western blot analyses revealed that both mRNA and protein expression of contractile smooth muscle markers were increased in isolated smooth muscle cells cultured under high compared with low glucose conditions. This effect was also observed in hyperglycemic Akita mice and in diabetic patients. Elevated glucose activated the protein kinase C and Rho/Rho-kinase signaling pathways and stimulated actin polymerization. Glucose-induced expression of contractile smooth muscle markers in cultured cells could be partially or completely repressed by inhibitors of advanced glycation end products, L-type calcium channels, protein kinase C, Rho-kinase, actin polymerization, and myocardin-related transcription factors. Furthermore, genetic ablation of the miR-143/145 cluster prevented the effects of glucose on smooth muscle marker expression. In conclusion, these data demonstrate a possible link between hyperglycemia and vascular disease states associated with smooth muscle contractility.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Aged
- Animals
- Atherosclerosis/enzymology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Contractile Proteins/agonists
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cytoskeletal Proteins/agonists
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Gene Expression Regulation
- Humans
- Male
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Protein Kinase C/chemistry
- Protein Kinase C/metabolism
- Signal Transduction
- rho GTP-Binding Proteins/agonists
- rho GTP-Binding Proteins/metabolism
- rho-Associated Kinases/chemistry
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Tran Thi Hien
- From the Departments of Experimental Medical Sciences and
| | | | - Diana Dahan
- From the Departments of Experimental Medical Sciences and
| | - Mari Ekman
- From the Departments of Experimental Medical Sciences and
| | - Mario Grossi
- From the Departments of Experimental Medical Sciences and
| | - Johan Sjögren
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Johan Nilsson
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Thomas Braun
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Thomas Boettger
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Eliana Garcia-Vaz
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | - Karin Stenkula
- From the Departments of Experimental Medical Sciences and
| | - Karl Swärd
- From the Departments of Experimental Medical Sciences and
| | - Maria F Gomez
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | | |
Collapse
|
27
|
Vatta M. Mitochondria, contractile apparatus, and ion channels in the failing myocardium: Special relationships or dangerous liaisons? Heart Rhythm 2016; 13:1140-1141. [PMID: 26820511 DOI: 10.1016/j.hrthm.2016.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Matteo Vatta
- Department of Medical and Molecular Genetics, and Krannert Institute of Cardiology and the Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
28
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
29
|
Moreau M, Néant I, Webb SE, Miller AL, Riou JF, Leclerc C. Ca(2+) coding and decoding strategies for the specification of neural and renal precursor cells during development. Cell Calcium 2015; 59:75-83. [PMID: 26744233 DOI: 10.1016/j.ceca.2015.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/07/2015] [Accepted: 12/11/2015] [Indexed: 01/03/2023]
Abstract
During embryogenesis, a rise in intracellular Ca(2+) is known to be a widespread trigger for directing stem cells towards a specific tissue fate, but the precise Ca(2+) signalling mechanisms involved in achieving these pleiotropic effects are still poorly understood. In this review, we compare the Ca(2+) signalling events that appear to be one of the first steps in initiating and regulating both neural determination (neural induction) and kidney development (nephrogenesis). We have highlighted the necessary and sufficient role played by Ca(2+) influx and by Ca(2+) transients in the determination and differentiation of pools of neural or renal precursors. We have identified new Ca(2+) target genes involved in neural induction and we showed that the same Ca(2+) early target genes studied are not restricted to neural tissue but are also present in other tissues, principally in the pronephros. In this review, we also described a mechanism whereby the transcriptional control of gene expression during neurogenesis and nephrogenesis might be directly controlled by Ca(2+) signalling. This mechanism involves members of the Kcnip family such that a change in their binding properties to specific DNA sites is a result of Ca(2+) binding to EF-hand motifs. The different functions of Ca(2+) signalling during these two events illustrate the versatility of Ca(2+) as a second messenger.
Collapse
Affiliation(s)
- Marc Moreau
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France
| | - Isabelle Néant
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France
| | - Sarah E Webb
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, People's Republic of China
| | - Andrew L Miller
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, People's Republic of China; MBL, Woods Hole, MA, USA
| | - Jean-François Riou
- Université Pierre et Marie Curie-Paris VI, Equipe "Signalisation et Morphogenèse", UMR7622-Biologie du Développement, 9, quai Saint-Bernard, 75005 Paris, France; CNRS, Equipe "Signalisation et Morphogenèse", UMR7622-Biologie du Développement, 9, quai Saint-Bernard, 75005 Paris, France
| | - Catherine Leclerc
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France.
| |
Collapse
|
30
|
Bhattachariya A, Dahan D, Ekman M, Boettger T, Braun T, Swärd K, Hellstrand P, Albinsson S. Spontaneous activity and stretch-induced contractile differentiation are reduced in vascular smooth muscle of miR-143/145 knockout mice. Acta Physiol (Oxf) 2015; 215:133-43. [PMID: 26052659 DOI: 10.1111/apha.12536] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/19/2014] [Accepted: 06/02/2015] [Indexed: 01/20/2023]
Abstract
AIM Stretch is essential for maintaining the contractile phenotype of vascular smooth muscle cells, and small non-coding microRNAs are known to be important in this process. Using a Dicer knockout model, we have previously reported that microRNAs are essential for stretch-induced differentiation and regulation of L-type calcium channel expression. The aim of this study was to investigate the importance of the smooth muscle-enriched miR-143/145 microRNA cluster for stretch-induced differentiation of the portal vein. METHODS Contractile force and depolarization-induced calcium influx were determined in portal veins from wild-type and miR-143/145 knockout mice. Stretch-induced contractile differentiation was investigated by determination of mRNA expression following organ culture for 24 h under longitudinal load by a hanging weight. RESULTS In the absence of miR-143/145, stretch-induced mRNA expression of contractile markers in the portal vein was reduced. This was associated with decreased amplitude of spontaneous activity and depolarization-induced contractile and intracellular calcium responses, while contractile responses to 5-HT were largely maintained. We found that these effects correlated with a reduced basal expression of the pore-forming subunit of L-type calcium channels and an increased expression of CaMKIIδ and the transcriptional repressor DREAM. CONCLUSION Our results suggest that the microRNA-143/145 cluster plays a role in maintaining stretch-induced contractile differentiation and calcium signalling in the portal vein. This may have important implications for the use of these microRNAs as therapeutic targets in vascular disease.
Collapse
Affiliation(s)
- A Bhattachariya
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - D Dahan
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Ekman
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - T Boettger
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - T Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - K Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - P Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - S Albinsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
31
|
Martinsen A, Dessy C, Morel N. Regulation of calcium channels in smooth muscle: new insights into the role of myosin light chain kinase. Channels (Austin) 2015; 8:402-13. [PMID: 25483583 DOI: 10.4161/19336950.2014.950537] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smooth muscle myosin light chain kinase (MLCK) plays a crucial role in artery contraction, which regulates blood pressure and blood flow distribution. In addition to this role, MLCK contributes to Ca(2+) flux regulation in vascular smooth muscle (VSM) and in non-muscle cells, where cytoskeleton has been suggested to help Ca(2+) channels trafficking. This conclusion is based on the use of pharmacological inhibitors of MLCK and molecular and cellular techniques developed to down-regulate the enzyme. Dissimilarities have been observed between cells and whole tissues, as well as between large conductance and small resistance arteries. A differential expression in MLCK and ion channels (either voltage-dependent Ca(2+) channels or non-selective cationic channels) could account for these observations, and is in line with the functional properties of the arteries. A potential involvement of MLCK in the pathways modulating Ca(2+) entry in VSM is described in the present review.
Collapse
Key Words
- CaM, calmodulin
- ER, endoplasmic reticulum
- MLCK, myosin light chain kinase
- Myosin light chain kinase
- ROC, receptor-operated Ca2+ (channel)
- SMC, smooth muscle cell
- SOC, store-operated Ca2+ (channel)
- SR, sarcoplasmic reticulum
- TRP
- TRP, transient receptor potential (channel)
- VOC, voltage-operated Ca2+ (channel)
- VSM, vascular smooth muscle
- VSMC, vascular smooth muscle cell
- [Ca2+]cyt, cytosolic Ca2+ concentration
- siRNA, small interfering RNA
- vascular smooth muscle
- voltage-dependent calcium channels
Collapse
Affiliation(s)
- A Martinsen
- a Cell physiology; IoNS; UCLouvain ; Brussels , Belgium
| | | | | |
Collapse
|
32
|
Sun Z, Cao X, Hu Z, Zhang L, Wang H, Zhou H, Li D, Zhang S, Xie M. MiR-103 inhibits osteoblast proliferation mainly through suppressing Cav1.2 expression in simulated microgravity. Bone 2015; 76:121-8. [PMID: 25868801 DOI: 10.1016/j.bone.2015.04.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/17/2015] [Accepted: 04/02/2015] [Indexed: 01/22/2023]
Abstract
Emerging evidence indicates that microRNAs (miRNAs) play important roles in modulating osteoblast function and bone formation. However, the influence of miRNA on osteoblast proliferation and the possible mechanisms underlying remain to be defined. In this study, we aimed to investigate whether miR-103 regulates osteoblast proliferation under simulated microgravity condition through regulating Cav1.2, the primary subunit of L-type voltage sensitive calcium channels (LTCCs). We first investigated the effect of simulated microgravity on osteoblast proliferation and the outcomes clearly demonstrated that the mechanical unloading inhibits MC3T3-E1 osteoblast-like cell proliferation. Using quantitative Real-Time PCR (qRT-PCR), we provided data showing that miR-103 was up-regulated in response to simulated microgravity. In addition, we observed that up-regulation of miR-103 inhibited and down-regulation of miR-103 promoted osteoblast proliferation under simulated microgravity condition. Furthermore, knocking-down or over-expressing miR-103, respectively, up- or down-regulated the level of Cav1.2 expression and LTCC currents, suggesting that miR-103 acts as an endogenous attenuator of Cav1.2 in osteoblasts under simulated microgravity condition. More importantly, we showed that the effect of miR-103 on osteoblast proliferation was diminished in simulated microgravity, when co-transfecting miR-103 mimic or inhibitor with Cav1.2 siRNA. Taken together, our data suggest that miR-103 inhibits osteoblast proliferation mainly through suppression of Cav1.2 expression under simulated microgravity condition. This work may provide a novel mechanism of microgravity-induced detrimental effects on osteoblast proliferation, identifying miR-103 as a novel possible therapeutic target in bone remodeling disorders in this mechanical unloading.
Collapse
Affiliation(s)
- Zhongyang Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Lianchang Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Han Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Hua Zhou
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Dongtao Li
- Center of Cardiology, Navy General Hospital, 100048 Beijing, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China.
| | - Manjiang Xie
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China.
| |
Collapse
|
33
|
Albinsson S, Bhattachariya A, Hellstrand P. Stretch-dependent smooth muscle differentiation in the portal vein-role of actin polymerization, calcium signaling, and microRNAs. Microcirculation 2015; 21:230-8. [PMID: 24238368 DOI: 10.1111/micc.12106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/09/2013] [Indexed: 11/28/2022]
Abstract
The mechanical forces acting on SMC in the vascular wall are known to regulate processes such as vascular remodeling and contractile differentiation. However, investigations to elucidate the underlying mechanisms of mechanotransduction in smooth muscle have been hampered by technical limitations associated with mechanical studies on pressurized small arteries, due primarily to the small amount of available tissue. The murine portal vein is a relatively large vessel showing myogenic tone that in many respects recapitulates the properties of small resistance vessels. Studies on stretched portal veins to elucidate mechanisms of mechanotransduction in the vascular wall have shown that stretch-sensitive regulation of contractile differentiation is mediated via Rho-activation and actin polymerization, while stretch-induced growth is regulated by the MAPK pathway. In this review, we have summarized findings on mechanotransduction in the portal vein with focus on stretch-induced contractile differentiation and the role of calcium, actin polymerization and miRNAs in this response.
Collapse
|
34
|
Mattiazzi A, Bassani RA, Escobar AL, Palomeque J, Valverde CA, Vila Petroff M, Bers DM. Chasing cardiac physiology and pathology down the CaMKII cascade. Am J Physiol Heart Circ Physiol 2015; 308:H1177-91. [PMID: 25747749 DOI: 10.1152/ajpheart.00007.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/16/2015] [Indexed: 11/22/2022]
Abstract
Calcium dynamics is central in cardiac physiology, as the key event leading to the excitation-contraction coupling (ECC) and relaxation processes. The primary function of Ca(2+) in the heart is the control of mechanical activity developed by the myofibril contractile apparatus. This key role of Ca(2+) signaling explains the subtle and critical control of important events of ECC and relaxation, such as Ca(2+) influx and SR Ca(2+) release and uptake. The multifunctional Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) is a signaling molecule that regulates a diverse array of proteins involved not only in ECC and relaxation but also in cell death, transcriptional activation of hypertrophy, inflammation, and arrhythmias. CaMKII activity is triggered by an increase in intracellular Ca(2+) levels. This activity can be sustained, creating molecular memory after the decline in Ca(2+) concentration, by autophosphorylation of the enzyme, as well as by oxidation, glycosylation, and nitrosylation at different sites of the regulatory domain of the kinase. CaMKII activity is enhanced in several cardiac diseases, altering the signaling pathways by which CaMKII regulates the different fundamental proteins involved in functional and transcriptional cardiac processes. Dysregulation of these pathways constitutes a central mechanism of various cardiac disease phenomena, like apoptosis and necrosis during ischemia/reperfusion injury, digitalis exposure, post-acidosis and heart failure arrhythmias, or cardiac hypertrophy. Here we summarize significant aspects of the molecular physiology of CaMKII and provide a conceptual framework for understanding the role of the CaMKII cascade on Ca(2+) regulation and dysregulation in cardiac health and disease.
Collapse
Affiliation(s)
- Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina;
| | - Rosana A Bassani
- Centro de Engenharia Biomédica, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Ariel L Escobar
- Biological Engineering and Small Scale Technologies, School of Engineering, University of California, Merced, California; and
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Martín Vila Petroff
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, California
| |
Collapse
|
35
|
Ai X. SR calcium handling dysfunction, stress-response signaling pathways, and atrial fibrillation. Front Physiol 2015; 6:46. [PMID: 25745402 PMCID: PMC4333799 DOI: 10.3389/fphys.2015.00046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/30/2015] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. It is associated with a markedly increased risk of premature death due to embolic stroke and also complicates co-existing cardiovascular diseases such as heart failure. The prevalence of AF increases dramatically with age, and aging has been shown to be an independent risk of AF. Due to an aging population in the world, a growing body of AF patients are suffering a diminished quality of life and causing an associated economic burden. However, effective pharmacologic treatments and prevention strategies are lacking due to a poor understanding of the molecular and electrophysiologic mechanisms of AF in the failing and/or aged heart. Recent studies suggest that altered atrial calcium handling contributes to the onset and maintenance of AF. Here we review the role of stress-response kinases and calcium handling dysfunction in AF genesis in the aged and failing heart.
Collapse
Affiliation(s)
- Xun Ai
- Department of Cell and Molecular Physiology, Loyola University Chicago Maywood, IL, USA
| |
Collapse
|
36
|
Sun Z, Cao X, Zhang Z, Hu Z, Zhang L, Wang H, Zhou H, Li D, Zhang S, Xie M. Simulated microgravity inhibits L-type calcium channel currents partially by the up-regulation of miR-103 in MC3T3-E1 osteoblasts. Sci Rep 2015; 5:8077. [PMID: 25627864 PMCID: PMC4308706 DOI: 10.1038/srep08077] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/05/2015] [Indexed: 11/25/2022] Open
Abstract
L-type voltage-sensitive calcium channels (LTCCs), particularly Cav1.2 LTCCs, play fundamental roles in cellular responses to mechanical stimuli in osteoblasts. Numerous studies have shown that mechanical loading promotes bone formation, whereas the removal of this stimulus under microgravity conditions results in a reduction in bone mass. However, whether microgravity exerts an influence on LTCCs in osteoblasts and whether this influence is a possible mechanism underlying the observed bone loss remain unclear. In the present study, we demonstrated that simulated microgravity substantially inhibited LTCC currents and suppressed Cav1.2 at the protein level in MC3T3-E1 osteoblast-like cells. In addition, reduced Cav1.2 protein levels decreased LTCC currents in MC3T3-E1 cells. Moreover, simulated microgravity increased miR-103 expression. Cav1.2 expression and LTCC current densities both significantly increased in cells that were transfected with a miR-103 inhibitor under mechanical unloading conditions. These results suggest that simulated microgravity substantially inhibits LTCC currents in osteoblasts by suppressing Cav1.2 expression. Furthermore, the down-regulation of Cav1.2 expression and the inhibition of LTCCs caused by mechanical unloading in osteoblasts are partially due to miR-103 up-regulation. Our study provides a novel mechanism for microgravity-induced detrimental effects on osteoblasts, offering a new avenue to further investigate the bone loss induced by microgravity.
Collapse
Affiliation(s)
- Zhongyang Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Zhuo Zhang
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Lianchang Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Han Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Hua Zhou
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Dongtao Li
- Center of Cardiology, Navy General Hospital, 100048, Beijing, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| | - Manjiang Xie
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, Shaanxi, China
| |
Collapse
|
37
|
Eilers W, Jaspers RT, de Haan A, Ferrié C, Valdivieso P, Flück M. CaMKII content affects contractile, but not mitochondrial, characteristics in regenerating skeletal muscle. BMC PHYSIOLOGY 2014; 14:7. [PMID: 25515219 PMCID: PMC4277655 DOI: 10.1186/s12899-014-0007-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 10/23/2014] [Indexed: 11/21/2022]
Abstract
Background The multi-meric calcium/calmodulin-dependent protein kinase II (CaMKII) is the main CaMK in skeletal muscle and its expression increases with endurance training. CaMK family members are implicated in contraction-induced regulation of calcium handling, fast myosin type IIA expression and mitochondrial biogenesis. The objective of this study was to investigate the role of an increased CaMKII content for the expression of the contractile and mitochondrial phenotype in vivo. Towards this end we attempted to co-express alpha- and beta-CaMKII isoforms in skeletal muscle and characterised the effect on the contractile and mitochondrial phenotype. Results Fast-twitch muscle m. gastrocnemius (GM) and slow-twitch muscle m. soleus (SOL) of the right leg of 3-month old rats were transfected via electro-transfer of injected expression plasmids for native α/β CaMKII. Effects were identified from the comparison to control-transfected muscles of the contralateral leg and non-transfected muscles. α/β CaMKII content in muscle fibres was 4-5-fold increased 7 days after transfection. The transfection rate was more pronounced in SOL than GM muscle (i.e. 12.6 vs. 3.5%). The overexpressed α/β CaMKII was functional as shown through increased threonine 287 phosphorylation of β-CaMKII after isometric exercise and down-regulated transcripts COXI, COXIV, SDHB after high-intensity exercise in situ. α/β CaMKII overexpression under normal cage activity accelerated excitation-contraction coupling and relaxation in SOL muscle in association with increased SERCA2, ANXV and fast myosin type IIA/X content but did not affect mitochondrial protein content. These effects were observed on a background of regenerating muscle fibres. Conclusion Elevated CaMKII content promotes a slow-to-fast type fibre shift in regenerating muscle but is not sufficient to stimulate mitochondrial biogenesis in the absence of an endurance stimulus.
Collapse
Affiliation(s)
- Wouter Eilers
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom.
| | - Richard T Jaspers
- Laboratory for Myology, MOVE Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Arnold de Haan
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom. .,Laboratory for Myology, MOVE Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Céline Ferrié
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | - Paola Valdivieso
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | - Martin Flück
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom. .,Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| |
Collapse
|
38
|
Edvinsson L, Povlsen GK, Ahnstedt H, Waldsee R. CaMKII inhibition with KN93 attenuates endothelin and serotonin receptor-mediated vasoconstriction and prevents subarachnoid hemorrhage-induced deficits in sensorimotor function. J Neuroinflammation 2014; 11:207. [PMID: 25498987 PMCID: PMC4269841 DOI: 10.1186/s12974-014-0207-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/24/2014] [Indexed: 02/04/2023] Open
Abstract
Background It has been suggested that transcriptional upregulation of cerebral artery contractile endothelin (ETB) and 5-hydroxytryptamine (5-HT1B) receptors play an important role in the development of late cerebral ischemia and increased vasoconstriction after subarachnoid hemorrhage (SAH). We tested the hypothesis that inhibition of calcium calmodulin-dependent protein kinase II (CaMKII) may reduce cerebral vasoconstriction mediated by endothelin and serotonin receptors and improve neurological outcome after experimental SAH. Methods SAH was induced in adult rats by injection of 250 μL autologous blood into the basal cisterns. The CaMKII activity in cerebral vessels was studied by Western blot and immunohistochemistry. The vasomotor responses of middle cerebral and basilar arteries were measured in a sensitive myograph system. The functional outcome was examined by the rotating pole test 2 and 3 days after SAH. Results SAH induced a rapid early increase in phosphorylated CaMKII protein at 1 h that was attenuated by cisternal administration of the CaMKII inhibitor KN93 (0.501 μg/kg) 45 min prior and immediately after SAH as evaluated by Western blot. Application of KN93 at 1 h and every 12 h post-SAH significantly reduced vascular CaMKII immunoreactivity at 72 h. In addition, contractile responses of cerebral arteries to endothelin-1 (ET-1) and 5-hydroxycarboxamide (5-CT) were increased at this time-point. KN93 treatment significantly attenuated the contraction induced by ET-1 and 5-CT. Importantly, treatment with the CaMKII inhibitor prevented SAH-induced deficits in neurological function, as evaluated by the rotating pole test, and similar sensorimotor scores were seen in sham-operated animals. Conclusions The present study has shown that SAH is associated with increased contractile responses to ET-1 and 5-CT in cerebral arteries and enhanced early activation of CaMKII. Treatment with the CaMKII inhibitor KN93 attenuated the contractile responses and prevented impaired sensorimotor function after SAH.
Collapse
Affiliation(s)
- Lars Edvinsson
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University and Lund University Hospital, Lund, SE-221 84, Sweden.
| | | | | | | |
Collapse
|
39
|
Brini M, Calì T, Ottolini D, Carafoli E. Neuronal calcium signaling: function and dysfunction. Cell Mol Life Sci 2014; 71:2787-814. [PMID: 24442513 PMCID: PMC11113927 DOI: 10.1007/s00018-013-1550-7] [Citation(s) in RCA: 500] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/15/2013] [Accepted: 12/30/2013] [Indexed: 01/07/2023]
Abstract
Calcium (Ca(2+)) is an universal second messenger that regulates the most important activities of all eukaryotic cells. It is of critical importance to neurons as it participates in the transmission of the depolarizing signal and contributes to synaptic activity. Neurons have thus developed extensive and intricate Ca(2+) signaling pathways to couple the Ca(2+) signal to their biochemical machinery. Ca(2+) influx into neurons occurs through plasma membrane receptors and voltage-dependent ion channels. The release of Ca(2+) from the intracellular stores, such as the endoplasmic reticulum, by intracellular channels also contributes to the elevation of cytosolic Ca(2+). Inside the cell, Ca(2+) is controlled by the buffering action of cytosolic Ca(2+)-binding proteins and by its uptake and release by mitochondria. The uptake of Ca(2+) in the mitochondrial matrix stimulates the citric acid cycle, thus enhancing ATP production and the removal of Ca(2+) from the cytosol by the ATP-driven pumps in the endoplasmic reticulum and the plasma membrane. A Na(+)/Ca(2+) exchanger in the plasma membrane also participates in the control of neuronal Ca(2+). The impaired ability of neurons to maintain an adequate energy level may impact Ca(2+) signaling: this occurs during aging and in neurodegenerative disease processes. The focus of this review is on neuronal Ca(2+) signaling and its involvement in synaptic signaling processes, neuronal energy metabolism, and neurotransmission. The contribution of altered Ca(2+) signaling in the most important neurological disorders will then be considered.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Tito Calì
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Denis Ottolini
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Ernesto Carafoli
- Venetian Institute for Molecular Medicine (VIMM), Via G.Orus, 2, 35129 Padua, Italy
| |
Collapse
|
40
|
Abstract
Evolution has exploited the chemical properties of Ca(2+), which facilitate its reversible binding to the sites of irregular geometry offered by biological macromolecules, to select it as a carrier of cellular signals. A number of proteins bind Ca(2+) to specific sites: those intrinsic to membranes play the most important role in the spatial and temporal regulation of the concentration and movements of Ca(2+) inside cells. Those which are soluble, or organized in non-membranous structures, also decode the Ca(2+) message to be then transmitted to the targets of its regulation. Since Ca(2+) controls the most important processes in the life of cells, it must be very carefully controlled within the cytoplasm, where most of the targets of its signaling function reside. Membrane channels (in the plasma membrane and in the organelles) mediate the entrance of Ca(2+) into the cytoplasm, ATPases, exchangers, and the mitochondrial Ca(2+) uptake system remove Ca(2+) from it. The concentration of Ca(2+) in the external spaces, which is controlled essentially by its dynamic exchanges in the bone system, is much higher than inside cells, and can, under conditions of pathology, generate a situation of dangerous internal Ca(2+) overload. When massive and persistent, the Ca(2+) overload culminates in the death of the cell. Subtle conditions of cellular Ca(2+) dyshomeostasis that affect individual systems that control Ca(2+), generate cell disease phenotypes that are particularly severe in tissues in which the signaling function of Ca(2+) has special importance, e.g., the nervous system.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Biology, University of Padova, Via U. Bassi 58/B, I-35131, Padova, Italy,
| | | | | | | |
Collapse
|
41
|
Vincent KP, McCulloch AD, Edwards AG. Toward a hierarchy of mechanisms in CaMKII-mediated arrhythmia. Front Pharmacol 2014; 5:110. [PMID: 24994983 PMCID: PMC4062880 DOI: 10.3389/fphar.2014.00110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) activity has been shown to contribute to arrhythmogenesis in a remarkably broad range of cardiac pathologies. Several of these involve significant structural and electrophysiologic remodeling, whereas others are due to specific channelopathies, and are not typically associated with arrhythmogenic changes to protein expression or cellular and tissue structure. The ability of CaMKII to contribute to arrhythmia across such a broad range of phenotypes suggests one of two interpretations regarding the role of CaMKII in cardiac arrhythmia: (1) some CaMKII-dependent mechanism is a common driver of arrhythmia irrespective of the specific etiology of the disease, or (2) these different etiologies expose different mechanisms by which CaMKII is capable of promoting arrhythmia. In this review, we dissect the available mechanistic evidence to explore these two possibilities and discuss how the various molecular actions of CaMKII promote arrhythmia in different pathophysiologic contexts.
Collapse
Affiliation(s)
- Kevin P Vincent
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA ; Department of Medicine, University of California San Diego La Jolla, CA, USA
| | - Andrew G Edwards
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA ; Institute for Experimental Medicine, Oslo University Hospital Ullevål Oslo, Norway ; Simula Research Laboratory Lysaker, Norway
| |
Collapse
|
42
|
Grubb S, Speerschneider T, Occhipinti D, Fiset C, Olesen SP, Thomsen MB, Calloe K. Loss of K+ currents in heart failure is accentuated in KChIP2 deficient mice. J Cardiovasc Electrophysiol 2014; 25:896-904. [PMID: 24678923 DOI: 10.1111/jce.12422] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/06/2014] [Accepted: 03/12/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION KV 4 together with KV Channel-Interacting Protein 2 (KChIP2) mediate the fast recovering transient outward potassium current (I(to,f)) in the heart. KChIP2 is downregulated in human heart failure (HF), potentially underlying the loss of I(to,f). We investigated remodeling associated with HF hypothesizing that KChIP2 plays a central role in the modulation of outward K(+) currents in HF. METHODS AND RESULTS HF was induced by aortic banding in wild-type (WT) and KChIP2 deficient (KChIP2(-/-)) mice, evaluated by echocardiography. Action potentials were measured by floating microelectrodes in intact hearts. Ventricular cardiomyocytes were isolated and whole-cell currents were recorded by patch clamp. Left ventricular action potentials in KChIP2(-/-) mice were prolonged in a rate dependent manner, consistent with patch-clamp data showing loss of a fast recovering outward K(+) current and upregulation of the slow recovering I(to,s) and I(Kur). HF decreased all outward K(+) currents in WT mice and did not change the relative contribution of I(to,f) in WT mice. Compared to WT HF, KChIP2(-/-) HF had a larger reduction of K(+) -current density. However, the relative APD prolongation caused by HF was shorter for KChIP2(-/-) compared with WT, and the APs of the 2 HF mouse types were indistinguishable. CONCLUSION I(to,f) is just one of many K(+) currents being downregulated in murine HF. The downregulation of repolarizing currents in HF is accentuated in KChIP2(-/-) mice. However, the prolongation of APs associated with HF is less in KChIP2(-/-) compared to WT, suggesting other compensatory mechanism(s) in the KChIP2(-/-) mouse.
Collapse
Affiliation(s)
- Søren Grubb
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tobias Speerschneider
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dona Occhipinti
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Céline Fiset
- Faculty of Pharmacy, Research Center of the Montreal Heart Institute, University of Montreal, Montreal, Quebec, Canada
| | - Søren-Peter Olesen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten B Thomsen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Calloe
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
McManus DD, Lin H, Tanriverdi K, Quercio M, Yin X, Larson MG, Ellinor PT, Levy D, Freedman JE, Benjamin EJ. Relations between circulating microRNAs and atrial fibrillation: data from the Framingham Offspring Study. Heart Rhythm 2014; 11:663-9. [PMID: 24444445 PMCID: PMC4219255 DOI: 10.1016/j.hrthm.2014.01.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND MicroRNA (miRNA) expression in atrial tissue has been implicated in pathologic susceptibility to atrial fibrillation (AF). Nevertheless, data on how circulating levels relate to AF are limited. OBJECTIVE The purpose of this study was to test the hypothesis that circulating miRNAs are associated with AF. METHODS Among 2445 Framingham Heart Study Offspring participants, we measured the expression of 385 circulating whole blood miRNAs by high-throughput quantitative reverse transcriptase polymerase chain reaction. We related miRNA levels with prevalent and new-onset AF. RESULTS Mean age of the cohort was 66.3 ± 8.9 years, and 56% were women; 153 participants had clinically apparent AF at baseline, and 107 developed AF during median follow-up of 5.4 years. miRNA-328 (miR-328) expression was lower among participants with prevalent AF (8.76 cycle threshold) compared to individuals with no AF (7.75 cycle threshold, P <.001). The association between miR-328 and prevalent AF persisted after adjustment for age, sex, and technical covariates (odds ratio 1.21, P = 1.8 × 10(-4)) but was attenuated in analyses adjusting for clinical AF risk factors (odds ratio 1.14, P = .017). In contrast to the associations between miR-328 and prevalent AF, none of the circulating miRNAs were associated with incident AF. CONCLUSION Circulating levels of miR-328, a miRNA known to promote atrial electrical remodeling by reducing L-type Ca(2+) channel density, were associated with prevalent AF. Adjustment for risk factors that promote atrial remodeling, including hypertension, attenuated the association between miR-328 and AF, potentially implicating miR-328 as a potential mediator of atrial remodeling and AF vulnerability.
Collapse
Affiliation(s)
- David D McManus
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Cardiology Division, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Epidemiology Division, Department of Quantitative Health Sciences, University of Massachusetts Medical School Worcester, Massachusetts.
| | - Honghuang Lin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Computational Biomedicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Kahraman Tanriverdi
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Cardiology Division, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michael Quercio
- Cardiology Division, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Xiaoyan Yin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts
| | - Martin G Larson
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Department of Mathematics and Statistics, Boston University, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Patrick T Ellinor
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, Massachusetts; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Daniel Levy
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Population Research Branch and Division of Intramural Research, National Heart, Lung, and Blood Institute of the National Institutes of Health, Bethesda, Massachusetts
| | - Jane E Freedman
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Cardiology Division, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Emelia J Benjamin
- Section of Cardiovascular Medicine, Department of Medicine, Boston University, Boston, Massachusetts; Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Epidemiology Department, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
44
|
Kreusser MM, Backs J. Integrated mechanisms of CaMKII-dependent ventricular remodeling. Front Pharmacol 2014; 5:36. [PMID: 24659967 PMCID: PMC3950490 DOI: 10.3389/fphar.2014.00036] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 12/20/2022] Open
Abstract
CaMKII has been shown to be activated during different cardiac pathological processes, and CaMKII-dependent mechanisms contribute to pathological cardiac remodeling, cardiac arrhythmias, and contractile dysfunction during heart failure. Activation of CaMKII during cardiac stress results in a broad number of biological effects such as, on the one hand, acute effects due to phosphorylation of distinct cellular proteins as ion channels and calcium handling proteins and, on the other hand, integrative mechanisms by changing gene expression. This review focuses on transcriptional and epigenetic effects of CaMKII activation during chronic cardiac remodeling. Multiple mechanisms have been described how CaMKII mediates changes in cardiac gene expression. CaMKII has been shown to directly phosphorylate components of the cardiac gene regulation machinery. CaMKII phosphorylates several transcription factors such as CREB that induces the activation of specific gene programs. CaMKII activates transcriptional regulators also indirectly by phosphorylating histone deacetylases, especially HDAC4, which in turn inhibits transcription factors that drive cardiac hypertrophy, fibrosis, and dysfunction. Recent studies demonstrate that CaMKII also phosphorylate directly histones, which may contribute to changes in gene expression. These findings of CaMKII-dependent gene regulation during cardiac remodeling processes suggest novel strategies for CaMKII-dependent “transcriptional or epigenetic therapies” to control cardiac gene expression and function. Manipulation of CaMKII-dependent signaling pathways in the settings of pathological cardiac growth, remodeling, and heart failure represents an auspicious therapeutic approach.
Collapse
Affiliation(s)
- Michael M Kreusser
- Research Unit Cardiac Epigenetics, Department of Cardiology, University of Heidelberg Heidelberg, Germany ; German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Germany
| | - Johannes Backs
- Research Unit Cardiac Epigenetics, Department of Cardiology, University of Heidelberg Heidelberg, Germany ; German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Germany
| |
Collapse
|
45
|
Schwoerer AP, Neef S, Broichhausen I, Jacubeit J, Tiburcy M, Wagner M, Biermann D, Didié M, Vettel C, Maier LS, Zimmermann WH, Carrier L, Eschenhagen T, Volk T, El-Armouche A, Ehmke H. Enhanced Ca²+ influx through cardiac L-type Ca²+ channels maintains the systolic Ca²+ transient in early cardiac atrophy induced by mechanical unloading. Pflugers Arch 2013; 465:1763-73. [PMID: 23842739 PMCID: PMC3898408 DOI: 10.1007/s00424-013-1316-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 11/04/2022]
Abstract
Cardiac atrophy as a consequence of mechanical unloading develops following exposure to microgravity or prolonged bed rest. It also plays a central role in the reverse remodelling induced by left ventricular unloading in patients with heart failure. Surprisingly, the intracellular Ca2+ transients which are pivotal to electromechanical coupling and to cardiac plasticity were repeatedly found to remain unaffected in early cardiac atrophy. To elucidate the mechanisms underlying the preservation of the Ca2+ transients, we investigated Ca2+ cycling in cardiomyocytes from mechanically unloaded (heterotopic abdominal heart transplantation) and control (orthotopic) hearts in syngeneic Lewis rats. Following 2 weeks of unloading, sarcoplasmic reticulum (SR) Ca2+ content was reduced by ~55 %. Atrophic cardiac myocytes also showed a much lower frequency of spontaneous diastolic Ca2+ sparks and a diminished systolic Ca2+ release, even though the expression of ryanodine receptors was increased by ~30 %. In contrast, current clamp recordings revealed prolonged action potentials in endocardial as well as epicardial myocytes which were associated with a two to fourfold higher sarcolemmal Ca2+ influx under action potential clamp. In addition, Cav1.2 subunits which form the pore of L-type Ca2+ channels (LTCC) were upregulated in atrophic myocardium. These data suggest that in early cardiac atrophy induced by mechanical unloading, an augmented sarcolemmal Ca2+ influx through LTCC fully compensates for a reduced systolic SR Ca2+ release to preserve the Ca2+ transient. This interplay involves an electrophysiological remodelling as well as changes in the expression of cardiac ion channels.
Collapse
Affiliation(s)
- A. P. Schwoerer
- Department of Cellular and Integrative Physiology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - S. Neef
- Department of Cardiology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
| | - I. Broichhausen
- Department of Cellular and Integrative Physiology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - J. Jacubeit
- Department of Cellular and Integrative Physiology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - M. Tiburcy
- Institute of Pharmacology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - M. Wagner
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - D. Biermann
- Department of Cardiovascular Surgery, Center for Cardiology and Cardiovascular Surgery, University Heart Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
| | - M. Didié
- Department of Cardiology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- Institute of Pharmacology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - C. Vettel
- Institute of Pharmacology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - L. S. Maier
- Department of Cardiology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - W. H. Zimmermann
- Institute of Pharmacology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - L. Carrier
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
- Inserm, U974; CNRS, UMR7215; UPMC UM76, Institut de Myologie, Paris, 75013 France
| | - T. Eschenhagen
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
| | - T. Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - A. El-Armouche
- Institute of Pharmacology, Heart Research Center, Georg-August-University Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research)—Goettingen, Goettingen, Germany
| | - H. Ehmke
- Department of Cellular and Integrative Physiology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research)—Hamburg/Kiel/Luebeck, Hamburg, Germany
| |
Collapse
|
46
|
Turczyńska KM, Hellstrand P, Swärd K, Albinsson S. Regulation of vascular smooth muscle mechanotransduction by microRNAs and L-type calcium channels. Commun Integr Biol 2013; 6:e22278. [PMID: 23802033 PMCID: PMC3689564 DOI: 10.4161/cib.22278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/17/2012] [Accepted: 09/18/2012] [Indexed: 11/19/2022] Open
Abstract
The phenotype of smooth muscle cells is regulated by multiple environmental factors including mechanical forces. Mechanical stretch of mouse portal veins ex vivo has been shown to promote contractile differentiation by activation of the Rho-pathway, an effect that is dependent on the influx of calcium via L-type calcium channels. MicroRNAs have recently been demonstrated to play a significant role in the control of smooth muscle phenotype and in a recent report we investigated their role in vascular mechanosensing. By smooth muscle specific deletion of Dicer, we found that microRNAs are essential for smooth muscle differentiation in response to stretch by regulating CamKIIδ and L-type calcium channel expression. Furthermore, we suggest that loss of L-type calcium channels in Dicer KO is due to reduced expression of the smooth muscle-enriched microRNA, miR-145, which targets CamKIIδ. These results unveil a novel mechanism for miR-145 dependent regulation of smooth muscle phenotype.
Collapse
|
47
|
Westenbrink BD, Edwards AG, McCulloch AD, Brown JH. The promise of CaMKII inhibition for heart disease: preventing heart failure and arrhythmias. Expert Opin Ther Targets 2013; 17:889-903. [PMID: 23789646 DOI: 10.1517/14728222.2013.809064] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Calcium-calmodulin-dependent protein kinase II (CaMKII) has emerged as a central mediator of cardiac stress responses which may serve several critical roles in the regulation of cardiac rhythm, cardiac contractility and growth. Sustained and excessive activation of CaMKII during cardiac disease has, however, been linked to arrhythmias, and maladaptive cardiac remodeling, eventually leading to heart failure (HF) and sudden cardiac death. AREAS COVERED In the current review, the authors describe the unique structural and biochemical properties of CaMKII and focus on its physiological effects in cardiomyocytes. Furthermore, they provide evidence for a role of CaMKII in cardiac pathologies, including arrhythmogenesis, myocardial ischemia and HF development. The authors conclude by discussing the potential for CaMKII as a target for inhibition in heart disease. EXPERT OPINION CaMKII provides a promising nodal point for intervention that may allow simultaneous prevention of HF progression and development of arrhythmias. For future studies and drug development there is a strong rationale for the development of more specific CaMKII inhibitors. In addition, an improved understanding of the differential roles of CaMKII subtypes is required.
Collapse
Affiliation(s)
- B Daan Westenbrink
- University of California, Department of Pharmacology, San Diego, La Jolla, CA, USA
| | | | | | | |
Collapse
|
48
|
While systolic cardiomyocyte function is preserved, diastolic myocyte function and recovery from acidosis are impaired in CaMKIIδ-KO mice. J Mol Cell Cardiol 2013; 59:107-16. [PMID: 23473775 DOI: 10.1016/j.yjmcc.2013.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/18/2013] [Indexed: 12/14/2022]
Abstract
OBJECTIVE CaMKII contributes to impaired contractility in heart failure by inducing SR Ca(2+)-leak. CaMKII-inhibition in the heart was suggested to be a novel therapeutic principle. Different CaMKII isoforms exist. Specifically targeting CaMKIIδ, the dominant isoform in the heart, could be of therapeutic potential without impairing other CaMKII isoforms. RATIONALE We investigated whether cardiomyocyte function is affected by isoform-specific knockout (KO) of CaMKIIδ under basal conditions and upon stress, i.e. upon ß-adrenergic stimulation and during acidosis. RESULTS Systolic cardiac function was largely preserved in the KO in vivo (echocardiography) corresponding to unchanged Ca(2+)-transient amplitudes and isolated myocyte contractility in vitro. CaMKII activity was dramatically reduced while phosphatase-1 inhibitor-1 was significantly increased. Surprisingly, while diastolic Ca(2+)-elimination was slower in KO most likely due to decreased phospholamban Thr-17 phosphorylation, frequency-dependent acceleration of relaxation was still present. Despite decreased SR Ca(2+)-reuptake at lower frequencies, SR Ca(2+)-content was not diminished, which might be due to reduced diastolic SR Ca(2+)-loss in the KO as a consequence of lower RyR Ser-2815 phosphorylation. Challenging KO myocytes with isoproterenol showed intact inotropic and lusitropic responses. During acidosis, SR Ca(2+)-reuptake and SR Ca(2+)-loading were significantly impaired in KO, resulting in an inability to maintain systolic Ca(2+)-transients during acidosis and impaired recovery. CONCLUSIONS Inhibition of CaMKIIδ appears to be safe under basal physiologic conditions. Specific conditions exist (e.g. during acidosis) under which CaMKII-inhibition might not be helpful or even detrimental. These conditions will have to be more clearly defined before CaMKII inhibition is used therapeutically.
Collapse
|
49
|
|
50
|
Naranjo JR, Mellström B. Ca2+-dependent transcriptional control of Ca2+ homeostasis. J Biol Chem 2012; 287:31674-80. [PMID: 22822058 DOI: 10.1074/jbc.r112.384982] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Intracellular free Ca(2+) ions regulate many cellular functions, and in turn, the cell devotes many genes/proteins to keep tight control of the level of intracellular free Ca(2+). Here, we review recent work on Ca(2+)-dependent mechanisms and effectors that regulate the transcription of genes encoding proteins involved in the maintenance of the homeostasis of Ca(2+) in the cell.
Collapse
Affiliation(s)
- Jose R Naranjo
- National Center of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC) and the Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.
| | | |
Collapse
|