51
|
Ning S, Hua L, Ji Z, Fan D, Meng X, Li Z, Wang Q, Guo Z. Protein 4.1 family and ion channel proteins interact to regulate the process of heart failure in rats. Acta Histochem 2021; 123:151748. [PMID: 34271280 DOI: 10.1016/j.acthis.2021.151748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a major cause of death in cardiovascular diseases worldwide, and its molecular mechanisms and effective prevention strategies remain to be further studied. The myocardial cytoskeleton plays a pivotal role in many heart diseases. However, little is known about the function of the membrane cytoskeleton 4.1 protein family and related regulatory mechanisms in the pathogenesis of HF. In this study, we detected the localization and expression of the protein 4.1 family and ion channel proteins in a rat HF model induced by doxorubicin (DOX), and studied the interactions between them. Our results showed that compared with the control group, the HF group displayed an increased expression level of protein 4.1R and decreased levels of protein 4.1 G and 4.1 N. The Nav1.5 protein levels were significantly increased, while the SERCA2a and Cav1.2 protein levels were significantly decreased in the HF group. Furthermore, there is co-localization and interaction between protein 4.1R and Nav1.5, protein 4.1 G and SERCA2a, protein 4.1 N and Cav1.2, respectively. Taken together, the results indicated that the protein 4.1 family might be involved in the occurrence and development of HF through its interaction with ion channel proteins, suggesting that 4.1 proteins may serve as a novel therapeutic target for HF.
Collapse
Affiliation(s)
- Shuwei Ning
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Lei Hua
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangguang Meng
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhiying Li
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Qian Wang
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhikun Guo
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
52
|
Grote Beverborg N, Später D, Knöll R, Hidalgo A, Yeh ST, Elbeck Z, Silljé HHW, Eijgenraam TR, Siga H, Zurek M, Palmér M, Pehrsson S, Albery T, Bomer N, Hoes MF, Boogerd CJ, Frisk M, van Rooij E, Damle S, Louch WE, Wang QD, Fritsche-Danielson R, Chien KR, Hansson KM, Mullick AE, de Boer RA, van der Meer P. Phospholamban antisense oligonucleotides improve cardiac function in murine cardiomyopathy. Nat Commun 2021; 12:5180. [PMID: 34462437 PMCID: PMC8405807 DOI: 10.1038/s41467-021-25439-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality worldwide, highlighting an urgent need for novel treatment options, despite recent improvements. Aberrant Ca2+ handling is a key feature of HF pathophysiology. Restoring the Ca2+ regulating machinery is an attractive therapeutic strategy supported by genetic and pharmacological proof of concept studies. Here, we study antisense oligonucleotides (ASOs) as a therapeutic modality, interfering with the PLN/SERCA2a interaction by targeting Pln mRNA for downregulation in the heart of murine HF models. Mice harboring the PLN R14del pathogenic variant recapitulate the human dilated cardiomyopathy (DCM) phenotype; subcutaneous administration of PLN-ASO prevents PLN protein aggregation, cardiac dysfunction, and leads to a 3-fold increase in survival rate. In another genetic DCM mouse model, unrelated to PLN (Cspr3/Mlp-/-), PLN-ASO also reverses the HF phenotype. Finally, in rats with myocardial infarction, PLN-ASO treatment prevents progression of left ventricular dilatation and improves left ventricular contractility. Thus, our data establish that antisense inhibition of PLN is an effective strategy in preclinical models of genetic cardiomyopathy as well as ischemia driven HF.
Collapse
Affiliation(s)
- Niels Grote Beverborg
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daniela Später
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden.
| | - Ralph Knöll
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Alejandro Hidalgo
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
- Murdoch Children's Research Institute (MCRI), Flemington, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | | | - Zaher Elbeck
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Herman H W Silljé
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim R Eijgenraam
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Humam Siga
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Magdalena Zurek
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Malin Palmér
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Laboratory of Experimental Biomedicine, Core Facilities, Sahlgrenska Academy, Gothenburg University, Göteborg, Sweden
| | - Susanne Pehrsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tamsin Albery
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nils Bomer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martijn F Hoes
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cornelis J Boogerd
- Department of Molecular Cardiology, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Eva van Rooij
- Department of Molecular Cardiology, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Regina Fritsche-Danielson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kenneth R Chien
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
- Department of Cell and Molecular Biology (CMB), Karolinska Institute, Stockholm, Sweden
| | - Kenny M Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Rudolf A de Boer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
53
|
Nothing Regular about the Regulins: Distinct Functional Properties of SERCA Transmembrane Peptide Regulatory Subunits. Int J Mol Sci 2021; 22:ijms22168891. [PMID: 34445594 PMCID: PMC8396278 DOI: 10.3390/ijms22168891] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
The sarco-endoplasmic reticulum calcium ATPase (SERCA) is responsible for maintaining calcium homeostasis in all eukaryotic cells by actively transporting calcium from the cytosol into the sarco-endoplasmic reticulum (SR/ER) lumen. Calcium is an important signaling ion, and the activity of SERCA is critical for a variety of cellular processes such as muscle contraction, neuronal activity, and energy metabolism. SERCA is regulated by several small transmembrane peptide subunits that are collectively known as the “regulins”. Phospholamban (PLN) and sarcolipin (SLN) are the original and most extensively studied members of the regulin family. PLN and SLN inhibit the calcium transport properties of SERCA and they are required for the proper functioning of cardiac and skeletal muscles, respectively. Myoregulin (MLN), dwarf open reading frame (DWORF), endoregulin (ELN), and another-regulin (ALN) are newly discovered tissue-specific regulators of SERCA. Herein, we compare the functional properties of the regulin family of SERCA transmembrane peptide subunits and consider their regulatory mechanisms in the context of the physiological and pathophysiological roles of these peptides. We present new functional data for human MLN, ELN, and ALN, demonstrating that they are inhibitors of SERCA with distinct functional consequences. Molecular modeling and molecular dynamics simulations of SERCA in complex with the transmembrane domains of MLN and ALN provide insights into how differential binding to the so-called inhibitory groove of SERCA—formed by transmembrane helices M2, M6, and M9—can result in distinct functional outcomes.
Collapse
|
54
|
Aguayo-Ortiz R, Creech J, Jiménez-Vázquez EN, Guerrero-Serna G, Wang N, da Rocha AM, Herron TJ, Espinoza-Fonseca LM. A multiscale approach for bridging the gap between potency, efficacy, and safety of small molecules directed at membrane proteins. Sci Rep 2021; 11:16580. [PMID: 34400719 PMCID: PMC8368179 DOI: 10.1038/s41598-021-96217-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/06/2021] [Indexed: 01/17/2023] Open
Abstract
Membrane proteins constitute a substantial fraction of the human proteome, thus representing a vast source of therapeutic drug targets. Indeed, newly devised technologies now allow targeting "undruggable" regions of membrane proteins to modulate protein function in the cell. Despite the advances in technology, the rapid translation of basic science discoveries into potential drug candidates targeting transmembrane protein domains remains challenging. We address this issue by harmonizing single molecule-based and ensemble-based atomistic simulations of ligand-membrane interactions with patient-derived induced pluripotent stem cell (iPSC)-based experiments to gain insights into drug delivery, cellular efficacy, and safety of molecules directed at membrane proteins. In this study, we interrogated the pharmacological activation of the cardiac Ca2+ pump (Sarcoplasmic reticulum Ca2+-ATPase, SERCA2a) in human iPSC-derived cardiac cells as a proof-of-concept model. The combined computational-experimental approach serves as a platform to explain the differences in the cell-based activity of candidates with similar functional profiles, thus streamlining the identification of drug-like candidates that directly target SERCA2a activation in human cardiac cells. Systematic cell-based studies further showed that a direct SERCA2a activator does not induce cardiotoxic pro-arrhythmogenic events in human cardiac cells, demonstrating that pharmacological stimulation of SERCA2a activity is a safe therapeutic approach targeting the heart. Overall, this novel multiscale platform encompasses organ-specific drug potency, efficacy, and safety, and opens new avenues to accelerate the bench-to-patient research aimed at designing effective therapies directed at membrane protein domains.
Collapse
Affiliation(s)
- Rodrigo Aguayo-Ortiz
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico, Mexico
| | - Jeffery Creech
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eric N Jiménez-Vázquez
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Guadalupe Guerrero-Serna
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nulang Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andre Monteiro da Rocha
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Todd J Herron
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
- CARTOX, Inc., 56655 Grand River Ave., PO Box 304, New Hudson, MI, 48165, USA
| | - L Michel Espinoza-Fonseca
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
55
|
Andjus S, Morillon A, Wery M. From Yeast to Mammals, the Nonsense-Mediated mRNA Decay as a Master Regulator of Long Non-Coding RNAs Functional Trajectory. Noncoding RNA 2021; 7:ncrna7030044. [PMID: 34449682 PMCID: PMC8395947 DOI: 10.3390/ncrna7030044] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/22/2022] Open
Abstract
The Nonsense-Mediated mRNA Decay (NMD) has been classically viewed as a translation-dependent RNA surveillance pathway degrading aberrant mRNAs containing premature stop codons. However, it is now clear that mRNA quality control represents only one face of the multiple functions of NMD. Indeed, NMD also regulates the physiological expression of normal mRNAs, and more surprisingly, of long non-coding (lnc)RNAs. Here, we review the different mechanisms of NMD activation in yeast and mammals, and we discuss the molecular bases of the NMD sensitivity of lncRNAs, considering the functional roles of NMD and of translation in the metabolism of these transcripts. In this regard, we describe several examples of functional micropeptides produced from lncRNAs. We propose that translation and NMD provide potent means to regulate the expression of lncRNAs, which might be critical for the cell to respond to environmental changes.
Collapse
Affiliation(s)
- Sara Andjus
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, PSL University, Sorbonne Université, CNRS UMR3244, 26 Rue d’Ulm, CEDEX 05, F-75248 Paris, France;
| | - Antonin Morillon
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, Sorbonne Université, CNRS UMR3244, 26 Rue d’Ulm, CEDEX 05, F-75248 Paris, France
- Correspondence: (A.M.); (M.W.)
| | - Maxime Wery
- ncRNA, Epigenetic and Genome Fluidity, Institut Curie, Sorbonne Université, CNRS UMR3244, 26 Rue d’Ulm, CEDEX 05, F-75248 Paris, France
- Correspondence: (A.M.); (M.W.)
| |
Collapse
|
56
|
Song N, Yang M, Zhang H, Yang SK. Intracellular Calcium Homeostasis and Kidney Disease. Curr Med Chem 2021; 28:3647-3665. [PMID: 33138745 DOI: 10.2174/0929867327666201102114257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
Kidney disease is a serious health problem that burdens our healthcare system. It is crucial to find the accurate pathogenesis of various types of kidney disease to provide guidance for precise therapies for patients suffering from these diseases. However, the exact molecular mechanisms underlying these diseases have not been fully understood. Disturbance of calcium homeostasis in renal cells plays a fundamental role in the development of various types of kidney disease, such as primary glomerular disease, diabetic nephropathy, acute kidney injury and polycystic kidney disease, through promoting cell proliferation, stimulating extracellular matrix accumulation, aggravating podocyte injury, disrupting cellular energetics as well as dysregulating cell survival and death dynamics. As a result, preventing the disturbance of calcium homeostasis in specific renal cells (such as tubular cells, podocytes and mesangial cells) is becoming one of the most promising therapeutic strategies in the treatment of kidney disease. The endoplasmic reticulum and mitochondria are two vital organelles in this process. Calcium ions cycle between the endoplasmic reticulum and mitochondria at the conjugation of these two organelles known as the mitochondria-associated endoplasmic reticulum membrane, maintaining calcium homeostasis. The pharmacologic modulation of cellular calcium homeostasis can be viewed as a novel therapeutic method for renal diseases. Here, we will introduce calcium homeostasis under physiological conditions and the disturbance of calcium homeostasis in kidney diseases. We will focus on the calcium homeostasis regulation in renal cells (including tubular cells, podocytes and mesangial cells), especially in the mitochondria- associated endoplasmic reticulum membranes of these renal cells.
Collapse
Affiliation(s)
- Na Song
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Shi-Kun Yang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
57
|
Fisher ME, Bovo E, Aguayo-Ortiz R, Cho EE, Pribadi MP, Dalton MP, Rathod N, Lemieux MJ, Espinoza-Fonseca LM, Robia SL, Zima AV, Young HS. Dwarf open reading frame (DWORF) is a direct activator of the sarcoplasmic reticulum calcium pump SERCA. eLife 2021; 10:65545. [PMID: 34075877 PMCID: PMC8203291 DOI: 10.7554/elife.65545] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/01/2021] [Indexed: 01/05/2023] Open
Abstract
The sarco-plasmic reticulum calcium pump (SERCA) plays a critical role in the contraction-relaxation cycle of muscle. In cardiac muscle, SERCA is regulated by the inhibitor phospholamban. A new regulator, dwarf open reading frame (DWORF), has been reported to displace phospholamban from SERCA. Here, we show that DWORF is a direct activator of SERCA, increasing its turnover rate in the absence of phospholamban. Measurement of in-cell calcium dynamics supports this observation and demonstrates that DWORF increases SERCA-dependent calcium reuptake. These functional observations reveal opposing effects of DWORF activation and phospholamban inhibition of SERCA. To gain mechanistic insight into SERCA activation, fluorescence resonance energy transfer experiments revealed that DWORF has a higher affinity for SERCA in the presence of calcium. Molecular modeling and molecular dynamics simulations provide a model for DWORF activation of SERCA, where DWORF modulates the membrane bilayer and stabilizes the conformations of SERCA that predominate during elevated cytosolic calcium.
Collapse
Affiliation(s)
- M'Lynn E Fisher
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Rodrigo Aguayo-Ortiz
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, United States
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Marsha P Pribadi
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Michael P Dalton
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Nishadh Rathod
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, United States
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
58
|
Mareedu S, Million ED, Duan D, Babu GJ. Abnormal Calcium Handling in Duchenne Muscular Dystrophy: Mechanisms and Potential Therapies. Front Physiol 2021; 12:647010. [PMID: 33897454 PMCID: PMC8063049 DOI: 10.3389/fphys.2021.647010] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by the loss of dystrophin. DMD is associated with muscle degeneration, necrosis, inflammation, fatty replacement, and fibrosis, resulting in muscle weakness, respiratory and cardiac failure, and premature death. There is no curative treatment. Investigations on disease-causing mechanisms offer an opportunity to identify new therapeutic targets to treat DMD. An abnormal elevation of the intracellular calcium (Cai2+) concentration in the dystrophin-deficient muscle is a major secondary event, which contributes to disease progression in DMD. Emerging studies have suggested that targeting Ca2+-handling proteins and/or mechanisms could be a promising therapeutic strategy for DMD. Here, we provide an updated overview of the mechanistic roles the sarcolemma, sarcoplasmic/endoplasmic reticulum, and mitochondria play in the abnormal and sustained elevation of Cai2+ levels and their involvement in DMD pathogenesis. We also discuss current approaches aimed at restoring Ca2+ homeostasis as potential therapies for DMD.
Collapse
Affiliation(s)
- Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Emily D Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States.,Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, United States
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
59
|
Vitorino R, Guedes S, Amado F, Santos M, Akimitsu N. The role of micropeptides in biology. Cell Mol Life Sci 2021; 78:3285-3298. [PMID: 33507325 PMCID: PMC11073438 DOI: 10.1007/s00018-020-03740-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Micropeptides are small polypeptides coded by small open-reading frames. Progress in computational biology and the analyses of large-scale transcriptomes and proteomes have revealed that mammalian genomes produce a large number of transcripts encoding micropeptides. Many of these have been previously annotated as long noncoding RNAs. The role of micropeptides in cellular homeostasis maintenance has been demonstrated. This review discusses different types of micropeptides as well as methods to identify them, such as computational approaches, ribosome profiling, and mass spectrometry.
Collapse
Affiliation(s)
- Rui Vitorino
- Departamento de Cirurgia E Fisiologia, Faculdade de Medicina da Universidade Do Porto, UnIC, Porto, Portugal.
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal.
| | - Sofia Guedes
- Departamento de Química, LAQV-REQUIMTE, Universidade de Aveiro, Aveiro, Portugal
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Francisco Amado
- Departamento de Química, LAQV-REQUIMTE, Universidade de Aveiro, Aveiro, Portugal
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Manuel Santos
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal
| | | |
Collapse
|
60
|
Zhihao L, Jingyu N, Lan L, Michael S, Rui G, Xiyun B, Xiaozhi L, Guanwei F. SERCA2a: a key protein in the Ca 2+ cycle of the heart failure. Heart Fail Rev 2021; 25:523-535. [PMID: 31701344 DOI: 10.1007/s10741-019-09873-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Calcium ion (Ca2+) cycle plays a crucial role in the contraction and relaxation of cardiomyocytes. The sarcoplasmic reticulum (SR) acts as an organelle for storing Ca2+, which mediated the release and re-uptake of Ca2+ during contraction and relaxation. Disorders of SR function lead to the dysfunction of Ca2+ cycle and myocardial cell function. The sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) acts as a subtype of SERCA expressed in the heart, which mediates the contraction of cardiomyocytes and Ca2+ in the cytoplasm to re-enter into the SR. The rate of uptake of Ca2+ by the SR determines the rate of myocardial relaxation. The regulation of SERCA2a activity controls the contractility and relaxation of the heart, affecting cardiac function. The expression and activity of SERCA2a are reduced in failing hearts. Gene therapy by increasing the expression of SERCA2a in the heart has been proven effective. In addition, SERCA2a is regulated by a variety of factors, including transmembrane micropeptides, protein kinases, and post-translational modifications (PTMs). In this review, we discuss the regulatory factors of SERCA2a and provide new insights into future treatments and the direction of heart failure research. In addition, gene therapy for SERCA2a has recently emerged as therapeutic option and hence will be discussed in this review.
Collapse
Affiliation(s)
- Liu Zhihao
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Ni Jingyu
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Li Lan
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Sarhene Michael
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Guo Rui
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Bian Xiyun
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Liu Xiaozhi
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Fan Guanwei
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China. .,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
61
|
Yap JQ, Seflova J, Sweazey R, Artigas P, Robia SL. FXYD proteins and sodium pump regulatory mechanisms. J Gen Physiol 2021; 153:211866. [PMID: 33688925 PMCID: PMC7953255 DOI: 10.1085/jgp.202012633] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
The sodium/potassium-ATPase (NKA) is the enzyme that establishes gradients of sodium and potassium across the plasma membrane. NKA activity is tightly regulated for different physiological contexts through interactions with single-span transmembrane peptides, the FXYD proteins. This diverse family of regulators has in common a domain containing a Phe-X-Tyr-Asp (FXYD) motif, two conserved glycines, and one serine residue. In humans, there are seven tissue-specific FXYD proteins that differentially modulate NKA kinetics as appropriate for each system, providing dynamic responsiveness to changing physiological conditions. Our understanding of how FXYD proteins contribute to homeostasis has benefitted from recent advances described in this review: biochemical and biophysical studies have provided insight into regulatory mechanisms, genetic models have uncovered remarkable complexity of FXYD function in integrated physiological systems, new posttranslational modifications have been identified, high-resolution structural studies have revealed new details of the regulatory interaction with NKA, and new clinical correlations have been uncovered. In this review, we address the structural determinants of diverse FXYD functions and the special roles of FXYDs in various physiological systems. We also discuss the possible roles of FXYDs in protein trafficking and regulation of non-NKA targets.
Collapse
Affiliation(s)
- John Q Yap
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL
| | - Jaroslava Seflova
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL
| | - Ryan Sweazey
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Pablo Artigas
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL
| |
Collapse
|
62
|
Yan Y, Tang R, Li B, Cheng L, Ye S, Yang T, Han YC, Liu C, Dong Y, Qu LH, Lui KO, Yang JH, Huang ZP. The cardiac translational landscape reveals that micropeptides are new players involved in cardiomyocyte hypertrophy. Mol Ther 2021; 29:2253-2267. [PMID: 33677093 PMCID: PMC8261087 DOI: 10.1016/j.ymthe.2021.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/11/2020] [Accepted: 03/02/2021] [Indexed: 11/27/2022] Open
Abstract
Hypertrophic growth of cardiomyocytes is one of the major compensatory responses in the heart after physiological or pathological stimulation. Protein synthesis enhancement, which is mediated by the translation of messenger RNAs, is one of the main features of cardiomyocyte hypertrophy. Although the transcriptome shift caused by cardiac hypertrophy induced by different stimuli has been extensively investigated, translatome dynamics in this cellular process has been less studied. Here, we generated a nucleotide-resolution translatome as well as transcriptome data from isolated primary cardiomyocytes undergoing hypertrophy. More than 10,000 open reading frames (ORFs) were detected from the deep sequencing of ribosome-protected fragments (Ribo-seq), which orchestrated the shift of the translatome in hypertrophied cardiomyocytes. Our data suggest that rather than increase the translational rate of ribosomes, the increased efficiency of protein synthesis in cardiomyocyte hypertrophy was attributable to an increased quantity of ribosomes. In addition, more than 100 uncharacterized short ORFs (sORFs) were detected in long noncoding RNA genes from Ribo-seq with potential of micropeptide coding. In a random test of 15 candidates, the coding potential of 11 sORFs was experimentally supported. Three micropeptides were identified to regulate cardiomyocyte hypertrophy by modulating the activities of oxidative phosphorylation, the calcium signaling pathway, and the mitogen-activated protein kinase (MAPK) pathway. Our study provides a genome-wide overview of the translational controls behind cardiomyocyte hypertrophy and demonstrates an unrecognized role of micropeptides in cardiomyocyte biology.
Collapse
Affiliation(s)
- Youchen Yan
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Rong Tang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Bin Li
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Liangping Cheng
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Shangmei Ye
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Yan-Chuang Han
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Chen Liu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China
| | - Liang-Hu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Jian-Hua Yang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China.
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China.
| |
Collapse
|
63
|
Schlesinger D, Elsässer SJ. Revisiting sORFs: overcoming challenges to identify and characterize functional microproteins. FEBS J 2021; 289:53-74. [PMID: 33595896 DOI: 10.1111/febs.15769] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/17/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
Short ORFs (sORFs), that is, occurrences of a start and stop codon within 100 codons or less, can be found in organisms of all domains of life, outnumbering annotated protein-coding ORFs by orders of magnitude. Even though functional proteins smaller than 100 amino acids are known, the coding potential of sORFs has often been overlooked, as it is not trivial to predict and test for functionality within the large number of sORFs. Recent advances in ribosome profiling and mass spectrometry approaches, together with refined bioinformatic predictions, have enabled a huge leap forward in this field and identified thousands of likely coding sORFs. A relatively low number of small proteins or microproteins produced from these sORFs have been characterized so far on the molecular, structural, and/or mechanistic level. These however display versatile and, in some cases, essential cellular functions, allowing for the exciting possibility that many more, previously unknown small proteins might be encoded in the genome, waiting to be discovered. This review will give an overview of the steadily growing microprotein field, focusing on eukaryotic small proteins. We will discuss emerging themes in the molecular action of microproteins, as well as advances and challenges in microprotein identification and characterization.
Collapse
Affiliation(s)
- Dörte Schlesinger
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Simon J Elsässer
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
64
|
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
|
65
|
The transmembrane peptide DWORF activates SERCA2a via dual mechanisms. J Biol Chem 2021; 296:100412. [PMID: 33581112 PMCID: PMC7988493 DOI: 10.1016/j.jbc.2021.100412] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The Ca-ATPase isoform 2a (SERCA2a) pumps cytosolic Ca2+ into the sarcoplasmic reticulum (SR) of cardiac myocytes, enabling muscle relaxation during diastole. Abnormally high cytosolic [Ca2+] is a central factor in heart failure, suggesting that augmentation of SERCA2a Ca2+ transport activity could be a promising therapeutic approach. SERCA2a is inhibited by the protein phospholamban (PLB), and a novel transmembrane peptide, dwarf open reading frame (DWORF), is proposed to enhance SR Ca2+ uptake and myocyte contractility by displacing PLB from binding to SERCA2a. However, establishing DWORF’s precise physiological role requires further investigation. In the present study, we developed cell-based FRET biosensor systems that can report on protein–protein interactions and structural changes in SERCA2a complexes with PLB and/or DWORF. To test the hypothesis that DWORF competes with PLB to occupy the SERCA2a-binding site, we transiently transfected DWORF into a stable HEK cell line expressing SERCA2a labeled with a FRET donor and PLB labeled with a FRET acceptor. We observed a significant decrease in FRET efficiency, consistent with a decrease in the fraction of SERCA2a bound to PLB. Surprisingly, we also found that DWORF also activates SERCA’s enzymatic activity directly in the absence of PLB at subsaturating calcium levels. Using site-directed mutagenesis, we generated DWORF variants that do not activate SERCA, thus identifying residues P15 and W22 as necessary for functional SERCA2a–DWORF interactions. This work advances our mechanistic understanding of the regulation of SERCA2a by small transmembrane proteins and sets the stage for future therapeutic development in heart failure research.
Collapse
|
66
|
The nuclear envelope protein Net39 is essential for muscle nuclear integrity and chromatin organization. Nat Commun 2021; 12:690. [PMID: 33514739 PMCID: PMC7846557 DOI: 10.1038/s41467-021-20987-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 12/24/2020] [Indexed: 01/14/2023] Open
Abstract
Lamins and transmembrane proteins within the nuclear envelope regulate nuclear structure and chromatin organization. Nuclear envelope transmembrane protein 39 (Net39) is a muscle nuclear envelope protein whose functions in vivo have not been explored. We show that mice lacking Net39 succumb to severe myopathy and juvenile lethality, with concomitant disruption in nuclear integrity, chromatin accessibility, gene expression, and metabolism. These abnormalities resemble those of Emery-Dreifuss muscular dystrophy (EDMD), caused by mutations in A-type lamins (LMNA) and other genes, like Emerin (EMD). We observe that Net39 is downregulated in EDMD patients, implicating Net39 in the pathogenesis of this disorder. Our findings highlight the role of Net39 at the nuclear envelope in maintaining muscle chromatin organization, gene expression and function, and its potential contribution to the molecular etiology of EDMD.
Collapse
|
67
|
Li H, Wang C, Li L, Li L. Skeletal muscle non-shivering thermogenesis as an attractive strategy to combat obesity. Life Sci 2021; 269:119024. [PMID: 33450257 DOI: 10.1016/j.lfs.2021.119024] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Obesity is a chronic disease derived from disequilibrium between energy intake and energy expenditure and evolving as a challenging epidemiological disease in the 21st century. It is urgently necessary to solve this issue by searching for effective strategies and safe drugs. Skeletal muscle could be a potential therapeutic target for the prevention and treatment of obesity and its associated complications due to non-shivering thermogenesis (NST) function. Skeletal muscle NST is based dominantly on futile sarcoplasmic reticulum Ca2+ ATPase (SERCA) pump cycling that leads to a rise in cytosolic Ca2+, increased adenosine triphosphate (ATP) hydrolysis and heat production. This review will highlight the mechanisms of skeletal muscle NST, including SLN mediated SERCA pump futile cycling, SR-mitochondrial crosstalk and increased mitochondrial biogenesis, and thermogenesis induced by uncoupling proteins 3 (UCP3). We then summarize natural products targeting the pathogenesis of obesity via skeletal muscle NST, offering new insights into pharmacotherapy and potential drug candidates to combat obesity.
Collapse
Affiliation(s)
- Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Section of Endocrinology, School of Medicine, Yale University, New Haven 06520, USA.
| | - Can Wang
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Lingqiao Li
- Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou 317306, PR China
| |
Collapse
|
68
|
Chemello F, Wang Z, Li H, McAnally JR, Liu N, Bassel-Duby R, Olson EN. Degenerative and regenerative pathways underlying Duchenne muscular dystrophy revealed by single-nucleus RNA sequencing. Proc Natl Acad Sci U S A 2020; 117:29691-29701. [PMID: 33148801 PMCID: PMC7703557 DOI: 10.1073/pnas.2018391117] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle disorder characterized by cycles of degeneration and regeneration of multinucleated myofibers and pathological activation of a variety of other muscle-associated cell types. The extent to which different nuclei within the shared cytoplasm of a myofiber may display transcriptional diversity and whether individual nuclei within a multinucleated myofiber might respond differentially to DMD pathogenesis is unknown. Similarly, the potential transcriptional diversity among nonmuscle cell types within dystrophic muscle has not been explored. Here, we describe the creation of a mouse model of DMD caused by deletion of exon 51 of the dystrophin gene, which represents a prevalent disease-causing mutation in humans. To understand the transcriptional abnormalities and heterogeneity associated with myofiber nuclei, as well as other mononucleated cell types that contribute to the muscle pathology associated with DMD, we performed single-nucleus transcriptomics of skeletal muscle of mice with dystrophin exon 51 deletion. Our results reveal distinctive and previously unrecognized myonuclear subtypes within dystrophic myofibers and uncover degenerative and regenerative transcriptional pathways underlying DMD pathogenesis. Our findings provide insights into the molecular underpinnings of DMD, controlled by the transcriptional activity of different types of muscle and nonmuscle nuclei.
Collapse
Affiliation(s)
- Francesco Chemello
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Zhaoning Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hui Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John R McAnally
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
69
|
Sarcolipin Exhibits Abundant RNA Transcription and Minimal Protein Expression in Horse Gluteal Muscle. Vet Sci 2020; 7:vetsci7040178. [PMID: 33202832 PMCID: PMC7711957 DOI: 10.3390/vetsci7040178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 01/02/2023] Open
Abstract
Ca2+ regulation in equine muscle is important for horse performance, yet little is known about this species-specific regulation. We reported recently that horse encode unique gene and protein sequences for the sarcoplasmic reticulum (SR) Ca2+-transporting ATPase (SERCA) and the regulatory subunit sarcolipin (SLN). Here we quantified gene transcription and protein expression of SERCA and its inhibitory peptides in horse gluteus, as compared to commonly-studied rabbit skeletal muscle. RNA sequencing and protein immunoblotting determined that horse gluteus expresses the ATP2A1 gene (SERCA1) as the predominant SR Ca2+-ATPase isoform and the SLN gene as the most-abundant SERCA inhibitory peptide, as also found in rabbit skeletal muscle. Equine muscle expresses an insignificant level of phospholamban (PLN), another key SERCA inhibitory peptide expressed commonly in a variety of mammalian striated muscles. Surprisingly in horse, the RNA transcript ratio of SLN-to-ATP2A1 is an order of magnitude higher than in rabbit, while the corresponding protein expression ratio is an order of magnitude lower than in rabbit. Thus, SLN is not efficiently translated or maintained as a stable protein in horse muscle, suggesting a non-coding role for supra-abundant SLN mRNA. We propose that the lack of SLN and PLN inhibition of SERCA activity in equine muscle is an evolutionary adaptation that potentiates Ca2+ cycling and muscle contractility in a prey species domestically selected for speed.
Collapse
|
70
|
Brusa R, Magri F, Bresolin N, Comi GP, Corti S. Noncoding RNAs in Duchenne and Becker muscular dystrophies: role in pathogenesis and future prognostic and therapeutic perspectives. Cell Mol Life Sci 2020; 77:4299-4313. [PMID: 32350552 PMCID: PMC11105074 DOI: 10.1007/s00018-020-03537-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Noncoding RNAs (ncRNAs), such as miRNAs and long noncoding RNAs, are key regulators of gene expression at the post-transcriptional level and represent promising therapeutic targets and biomarkers for several human diseases, including Duchenne and Becker muscular dystrophies (DMD/BMD). A role for ncRNAs in the pathogenesis of muscular dystrophies has been suggested, even if it is still incompletely understood. Here, we discuss current progress leading towards the clinical utility of ncRNAs for DMD/BMD. Long and short noncoding RNAs are differentially expressed in DMD/BMD and have a mechanism of action via targeting mRNAs. A subset of muscle-enriched miRNAs, the so-called myomiRs (miR-1, miR-133, and miR-206), are increased in the serum of patients with DMD and in dystrophin-defective animal models. Interestingly, myomiRs might be used as biomarkers, given that their levels can be corrected after dystrophin restoration in dystrophic mice. Remarkably, further evidence demonstrates that ncRNAs also play a role in dystrophin expression; thus, their modulations might represent a potential therapeutic strategy with the aim of upregulating the dystrophin protein in combination with other oligonucleotides/gene therapy approaches.
Collapse
Affiliation(s)
- Roberta Brusa
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Magri
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nereo Bresolin
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.
| |
Collapse
|
71
|
Li XL, Pongor L, Tang W, Das S, Muys BR, Jones MF, Lazar SB, Dangelmaier EA, Hartford CCR, Grammatikakis I, Hao Q, Sun Q, Schetter A, Martindale JL, Tang B, Jenkins LM, Robles AI, Walker RL, Ambs S, Chari R, Shabalina SA, Gorospe M, Hussain SP, Harris CC, Meltzer PS, Prasanth KV, Aladjem MI, Andresson T, Lal A. A small protein encoded by a putative lncRNA regulates apoptosis and tumorigenicity in human colorectal cancer cells. eLife 2020; 9:e53734. [PMID: 33112233 PMCID: PMC7673786 DOI: 10.7554/elife.53734] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 10/27/2020] [Indexed: 12/17/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are often associated with polysomes, indicating coding potential. However, only a handful of endogenous proteins encoded by putative lncRNAs have been identified and assigned a function. Here, we report the discovery of a putative gastrointestinal-tract-specific lncRNA (LINC00675) that is regulated by the pioneer transcription factor FOXA1 and encodes a conserved small protein of 79 amino acids which we termed FORCP (FOXA1-Regulated Conserved Small Protein). FORCP transcript is undetectable in most cell types but is abundant in well-differentiated colorectal cancer (CRC) cells where it functions to inhibit proliferation, clonogenicity, and tumorigenesis. The epitope-tagged and endogenous FORCP protein predominantly localizes to the endoplasmic reticulum (ER). In response to ER stress, FORCP depletion results in decreased apoptosis. Our findings on the initial characterization of FORCP demonstrate that FORCP is a novel, conserved small protein encoded by a mis-annotated lncRNA that regulates apoptosis and tumorigenicity in well-differentiated CRC cells.
Collapse
Affiliation(s)
- Xiao Ling Li
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Lőrinc Pongor
- Developmental Therapeutics Branch, CCR, NCI, NIHBethesdaUnited States
| | - Wei Tang
- Molecular Epidemiology Section, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Sudipto Das
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, IncFrederickUnited States
| | - Bruna R Muys
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Matthew F Jones
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Sarah B Lazar
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Emily A Dangelmaier
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Corrine CR Hartford
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Ioannis Grammatikakis
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| | - Qinyu Hao
- Department of Cell and Developmental Biology, Cancer Center at Illinois University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Qinyu Sun
- Department of Cell and Developmental Biology, Cancer Center at Illinois University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Aaron Schetter
- Molecular Genetics and Carcinogenesis Section, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIHBaltimoreUnited States
| | - BinWu Tang
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIHBethesdaUnited States
| | - Lisa M Jenkins
- Laboratory of Cell Biology, CCR, NCI, NIHBethesdaUnited States
| | - Ana I Robles
- Molecular Genetics and Carcinogenesis Section, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Robert L Walker
- Molecular Genetics Section, Genetics Branch, CCR, NCI, NIHBethesdaUnited States
| | - Stefan Ambs
- Molecular Epidemiology Section, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, National Cancer InstituteFrederickUnited States
| | - Svetlana A Shabalina
- National Center for Biotechnology Information, National Library of Medicine, NIHBethesdaUnited States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIHBaltimoreUnited States
| | - S Perwez Hussain
- Pancreatic Cancer Unit, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Curtis C Harris
- Molecular Genetics and Carcinogenesis Section, Laboratory of Human Carcinogenesis, CCR, NCI, NIHBethesdaUnited States
| | - Paul S Meltzer
- Molecular Genetics Section, Genetics Branch, CCR, NCI, NIHBethesdaUnited States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer Center at Illinois University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, CCR, NCI, NIHBethesdaUnited States
| | - Thorkell Andresson
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, IncFrederickUnited States
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaUnited States
| |
Collapse
|
72
|
Braun JL, Geromella MS, Hamstra SI, Fajardo VA. Neuronatin regulates whole-body metabolism: is thermogenesis involved? FASEB Bioadv 2020; 2:579-586. [PMID: 33089074 PMCID: PMC7566048 DOI: 10.1096/fba.2020-00052] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
Neuronatin (NNAT) was originally discovered in 1995 and labeled as a brain developmental gene due to its abundant expression in developing brains. Over the past 25 years, researchers have uncovered NNAT in other tissues; notably, the hypothalamus, pancreatic β‐cells, and adipocytes. Recent evidence in these tissues indicates that NNAT plays a significant role in metabolism whereby it regulates food intake, insulin secretion, and adipocyte differentiation. Furthermore, genetic deletion of Nnat in mice lowers whole‐body energy expenditure and increases susceptibility to diet‐induced obesity and glucose intolerance; however, the underlying cellular mechanisms remain unknown. Based on its sequence homology with phospholamban, NNAT has a purported role in regulating the sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump. However, NNAT also shares sequence homology with sarcolipin, which has the unique property of uncoupling the SERCA pump, increasing whole‐body energy expenditure and thus promoting adaptive thermogenesis in states of caloric excess or cold exposure. Thus, in this article, we discuss the accumulating evidence suggestive of NNAT’s role in whole‐body metabolic regulation, while highlighting its potential to mediate adaptive thermogenesis via SERCA uncoupling.
Collapse
Affiliation(s)
- Jessica L Braun
- Department of Kinesiology Brock University St. Catharines ON USA.,Centre for Bone and Muscle Health Brock University St. Catharines ON USA.,Centre for Neuroscience Brock University St. Catharines ON USA
| | - Mia S Geromella
- Department of Kinesiology Brock University St. Catharines ON USA.,Centre for Bone and Muscle Health Brock University St. Catharines ON USA
| | - Sophie I Hamstra
- Department of Kinesiology Brock University St. Catharines ON USA.,Centre for Bone and Muscle Health Brock University St. Catharines ON USA
| | - Val A Fajardo
- Department of Kinesiology Brock University St. Catharines ON USA.,Centre for Bone and Muscle Health Brock University St. Catharines ON USA.,Centre for Neuroscience Brock University St. Catharines ON USA
| |
Collapse
|
73
|
Makarewich CA, Bezprozvannaya S, Gibson AM, Bassel-Duby R, Olson EN. Gene Therapy With the DWORF Micropeptide Attenuates Cardiomyopathy in Mice. Circ Res 2020; 127:1340-1342. [PMID: 32878549 DOI: 10.1161/circresaha.120.317156] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Catherine A Makarewich
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center (C.A.M., A.M.G.).,Pediatrics, University of Cincinnati College of Medicine (C.A.M.)
| | - Svetlana Bezprozvannaya
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center (C.A.M., A.M.G.)
| | - Aaron M Gibson
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center (C.A.M., A.M.G.)
| | - Rhonda Bassel-Duby
- Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center (S.B., R.B.-D., E.N.O.)
| | - Eric N Olson
- Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center (S.B., R.B.-D., E.N.O.)
| |
Collapse
|
74
|
Hamstra SI, Whitley KC, Baranowski RW, Kurgan N, Braun JL, Messner HN, Fajardo VA. The role of phospholamban and GSK3 in regulating rodent cardiac SERCA function. Am J Physiol Cell Physiol 2020; 319:C694-C699. [PMID: 32755452 DOI: 10.1152/ajpcell.00318.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac contractile function is largely mediated by the regulation of Ca2+ cycling throughout the lifespan. The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is paramount to cardiac Ca2+ regulation, and it is well established that SERCA dysfunction pathologically contributes to cardiomyopathy and heart failure. Phospholamban (PLN) is a well-known inhibitor of the SERCA pump and its regulation of SERCA2a-the predominant cardiac SERCA isoform-contributes significantly to proper cardiac function. Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase involved in several metabolic pathways, and we and others have shown that it regulates SERCA function. In this mini-review, we highlight the underlying mechanisms behind GSK3's regulation of SERCA function specifically discussing changes in SERCA2a and PLN expression and its potential protection against oxidative stress. Ultimately, these recent findings that we discuss could have clinical implications in the treatment and prevention of cardiomyopathies and heart failure.
Collapse
Affiliation(s)
- Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Kennedy C Whitley
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Holt N Messner
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
75
|
Sarcoplasmic reticulum calcium mishandling: central tenet in heart failure? Biophys Rev 2020; 12:865-878. [PMID: 32696300 DOI: 10.1007/s12551-020-00736-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Excitation-contraction coupling links excitation of the sarcolemmal surface membrane to mechanical contraction. In the heart this link is established via a Ca2+-induced Ca2+ release process, which, following sarcolemmal depolarisation, prompts Ca2+ release from the sarcoplasmic reticulum (SR) though the ryanodine receptor (RyR2). This substantially raises the cytoplasmic Ca2+ concentration to trigger systole. In diastole, Ca2+ is removed from the cytoplasm, primarily via the sarcoplasmic-endoplasmic reticulum Ca2+-dependent ATPase (SERCA) pump on the SR membrane, returning Ca2+ to the SR store. Ca2+ movement across the SR is thus fundamental to the systole/diastole cycle and plays an essential role in maintaining cardiac contractile function. Altered SR Ca2+ homeostasis (due to disrupted Ca2+ release, storage, and reuptake pathways) is a central tenet of heart failure and contributes to depressed contractility, impaired relaxation, and propensity to arrhythmia. This review will focus on the molecular mechanisms that underlie asynchronous Ca2+ cycling around the SR in the failing heart. Further, this review will illustrate that the combined effects of expression changes and disruptions to RyR2 and SERCA2a regulatory pathways are critical to the pathogenesis of heart failure.
Collapse
|
76
|
Aguayo-Ortiz R, Espinoza-Fonseca LM. Linking Biochemical and Structural States of SERCA: Achievements, Challenges, and New Opportunities. Int J Mol Sci 2020; 21:ijms21114146. [PMID: 32532023 PMCID: PMC7313052 DOI: 10.3390/ijms21114146] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Sarcoendoplasmic reticulum calcium ATPase (SERCA), a member of the P-type ATPase family of ion and lipid pumps, is responsible for the active transport of Ca2+ from the cytoplasm into the sarcoplasmic reticulum lumen of muscle cells, into the endoplasmic reticulum (ER) of non-muscle cells. X-ray crystallography has proven to be an invaluable tool in understanding the structural changes of SERCA, and more than 70 SERCA crystal structures representing major biochemical states (defined by bound ligand) have been deposited in the Protein Data Bank. Consequently, SERCA is one of the best characterized components of the calcium transport machinery in the cell. Emerging approaches in the field, including spectroscopy and molecular simulation, now help integrate and interpret this rich structural information to understand the conformational transitions of SERCA that occur during activation, inhibition, and regulation. In this review, we provide an overview of the crystal structures of SERCA, focusing on identifying metrics that facilitate structure-based categorization of major steps along the catalytic cycle. We examine the integration of crystallographic data with different biophysical approaches and computational methods to link biochemical and structural states of SERCA that are populated in the cell. Finally, we discuss the challenges and new opportunities in the field, including structural elucidation of functionally important and novel regulatory complexes of SERCA, understanding the structural basis of functional divergence among homologous SERCA regulators, and bridging the gap between basic and translational research directed toward therapeutic modulation of SERCA.
Collapse
|
77
|
The micropeptide LEMP plays an evolutionarily conserved role in myogenesis. Cell Death Dis 2020; 11:357. [PMID: 32393776 PMCID: PMC7214441 DOI: 10.1038/s41419-020-2570-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 01/16/2023]
Abstract
In recent years, micropeptides have been increasingly identified as important regulators in various biological processes. However, whether micropeptides are functionally conserved remains largely unknown. Here, we uncovered a micropeptide with evolutionarily conserved roles in myogenesis. RNA-seq data analysis of proliferating mouse satellite cells (SCs) and differentiated myotubes identified a previously annotated lncRNA, MyolncR4 (1500011K16RIK), which is upregulated during muscle differentiation. Significantly, MyolncR4 is highly conserved across vertebrate species. Multiple lines of evidence demonstrate that MyolncR4 encodes a 56-aa micropeptide, which was named as LEMP (lncRNA encoded micropeptide). LEMP promotes muscle formation and regeneration in mouse. In zebrafish, MyolncR4 is enriched in developing somites and elimination of LEMP results in impaired muscle development, which could be efficiently rescued by expression of the mouse LEMP. Interestingly, LEMP is localized at both the plasma membrane and mitochondria, and associated with multiple mitochondrial proteins, suggestive of its involvement in mitochondrial functions. Together, our work uncovers a micropeptide that plays an evolutionarily conserved role in skeletal muscle differentiation, pinpointing the functional importance of this growing family of small peptides.
Collapse
|
78
|
Dangelmaier E, Lal A. Adaptor proteins in long noncoding RNA biology. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194370. [DOI: 10.1016/j.bbagrm.2019.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
|
79
|
Hamstra SI, Kurgan N, Baranowski RW, Qiu L, Watson CJF, Messner HN, MacPherson REK, MacNeil AJ, Roy BD, Fajardo VA. Low-dose lithium feeding increases the SERCA2a-to-phospholamban ratio, improving SERCA function in murine left ventricles. Exp Physiol 2020; 105:666-675. [PMID: 32087034 DOI: 10.1113/ep088061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/21/2020] [Indexed: 12/24/2022]
Abstract
NEW FINDINGS What is the central question of this study? Inhibition of glycogen synthase kinase-3 (GSK3) has been shown to improve cardiac SERCA2a function. Lithium can inhibit GSK3, but therapeutic doses used in treating bipolar disorder can have toxic effects. It has not been determined whether subtherapeutic doses of lithium can improve cardiac SERCA function. What is the main finding and its importance? Using left ventricles from wild-type mice, we found that subtherapeutic lithium feeding for 6 weeks decreased GSK3 activity and increased cardiac SERCA function compared with control-fed mice. These findings warrant the investigation of low-dose lithium feeding in preclinical models of cardiomyopathy and heart failure to determine the therapeutic benefit of GSK3 inhibition. ABSTRACT The sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) pump is responsible for regulating calcium (Ca2+ ) within myocytes, with SERCA2a being the dominant isoform in cardiomyocytes. Its inhibitor, phospholamban (PLN), acts by decreasing the affinity of SERCA for Ca2+ . Changes in the SERCA2a:PLN ratio can cause Ca2+ dysregulation often seen in patients with dilated cardiomyopathy and heart failure. The enzyme glycogen synthase kinase-3 (GSK3) is known to downregulate SERCA function by decreasing the SERCA2a:PLN ratio. In this study, we sought to determine whether feeding mice low-dose lithium, a natural GSK3 inhibitor, would improve left ventricular SERCA function by altering the SERCA2a:PLN ratio. To this end, male wild-type C57BL/6J mice were fed low-dose lithium via drinking water (10 mg kg-1 day-1 LiCl for 6 weeks) and left ventricles were harvested. GSK3 activity was significantly reduced in LiCl-fed versus control-fed mice. The apparent affinity of SERCA for Ca2+ was also increased (pCa50 ; control, 6.09 ± 0.03 versus LiCl, 6.26 ± 0.04, P < 0.0001) along with a 2.0-fold increase in SERCA2a:PLN ratio in LiCl-fed versus control-fed mice. These findings suggest that low-dose lithium supplementation can improve SERCA function by increasing the SERCA2a:PLN ratio. Future studies in murine preclinical models will determine whether GSK3 inhibition via low-dose lithium could be a potential therapeutic strategy for dilated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Liqun Qiu
- Department of Chemistry, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Colton J F Watson
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Holt N Messner
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | | | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Brian D Roy
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| |
Collapse
|
80
|
Chen X, Zhang X, Gross S, Houser SR, Soboloff J. Acetylation of SERCA2a, Another Target for Heart Failure Treatment? Circ Res 2020; 124:1285-1287. [PMID: 31021723 DOI: 10.1161/circresaha.119.315017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xiongwen Chen
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Xiaoying Zhang
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Scott Gross
- the Fels Institute for Cancer Research and Molecular Biology (S.G., J.S.), Temple University School of Medicine, Philadelphia, PA
| | - Steven R Houser
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Jonathan Soboloff
- the Fels Institute for Cancer Research and Molecular Biology (S.G., J.S.), Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
81
|
Chen J, Sitsel A, Benoy V, Sepúlveda MR, Vangheluwe P. Primary Active Ca 2+ Transport Systems in Health and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035113. [PMID: 31501194 DOI: 10.1101/cshperspect.a035113] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calcium ions (Ca2+) are prominent cell signaling effectors that regulate a wide variety of cellular processes. Among the different players in Ca2+ homeostasis, primary active Ca2+ transporters are responsible for keeping low basal Ca2+ levels in the cytosol while establishing steep Ca2+ gradients across intracellular membranes or the plasma membrane. This review summarizes our current knowledge on the three types of primary active Ca2+-ATPases: the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pumps, the secretory pathway Ca2+- ATPase (SPCA) isoforms, and the plasma membrane Ca2+-ATPase (PMCA) Ca2+-transporters. We first discuss the Ca2+ transport mechanism of SERCA1a, which serves as a reference to describe the Ca2+ transport of other Ca2+ pumps. We further highlight the common and unique features of each isoform and review their structure-function relationship, expression pattern, regulatory mechanisms, and specific physiological roles. Finally, we discuss the increasing genetic and in vivo evidence that links the dysfunction of specific Ca2+-ATPase isoforms to a broad range of human pathologies, and highlight emerging therapeutic strategies that target Ca2+ pumps.
Collapse
Affiliation(s)
- Jialin Chen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Aljona Sitsel
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Veronick Benoy
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
82
|
Mbikou P, Rademaker MT, Charles CJ, Richards MA, Pemberton CJ. Cardiovascular effects of DWORF (dwarf open reading frame) peptide in normal and ischaemia/reperfused isolated rat hearts. Peptides 2020; 124:170192. [PMID: 31712056 DOI: 10.1016/j.peptides.2019.170192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/08/2019] [Accepted: 10/31/2019] [Indexed: 11/16/2022]
Abstract
The novel peptide dwarf open reading frame (DWORF), highly conserved across species and expressed almost exclusively in cardiac ventricular muscle, may play a role in cardiac physiology and pathophysiology. The effect of direct administration of DWORF in the intact heart has not previously been examined. Accordingly, we investigated the cardiac effects of DWORF (1-30 nM) in normal isolated perfused rat hearts and hearts undergoing ischaemia/reperfusion (I/R) injury, and evaluated potential mechanisms of action. Exogenous DWORF at the top dose (30 nM) increased perfusion pressure (PP) in normal hearts, which indicates coronary vasoconstriction; and during post-ischaemic reperfusion, DWORF increased PP in a dose-dependent manner. In I/R hearts, DWORF at the top dose also increased left ventricular end-diastolic pressure and maximum and minimum derivatives of left ventricular pressure noted dP/dt(max) and dP/dt(min), respectively, without affecting developed pressure (DP). Co-infusion of DWORF with Diltiazem, an l-type Ca2+ channel blocker (1μM), in I/R hearts attenuated the falls in DP, dP/dt(max) and dP/dt(min) observed with Diltiazem alone. DWORF co-infusion with both Diltiazem and Y27632 (1μM) (a Rho-Kinase inhibitor) reversed the coronary vasodilator effect of the inhibitors administered alone. In conclusion, we provide the first evidence that DWORF has coronary vasoconstrictor actions in normal hearts and when administered during reperfusion in an ex-vivo model of cardiac I/R injury, and also exhibits positive cardiac inotropic activity in the latter setting. DWORF's effect on ventricular contractile function appears to be dependent on the l-type Ca2+ channel, whereas Rho-Kinase activity may be related to the coronary vasoconstrictor effects of DWORF.
Collapse
Affiliation(s)
- Prisca Mbikou
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand.
| | - Miriam T Rademaker
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - Christopher J Charles
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - Mark A Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - Christopher J Pemberton
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| |
Collapse
|
83
|
Tang X, Pan L, Zhao S, Dai F, Chao M, Jiang H, Li X, Lin Z, Huang Z, Meng G, Wang C, Chen C, Liu J, Wang X, Ferro A, Wang H, Chen H, Gao Y, Lu Q, Xie L, Han Y, Ji Y. SNO-MLP (S-Nitrosylation of Muscle LIM Protein) Facilitates Myocardial Hypertrophy Through TLR3 (Toll-Like Receptor 3)-Mediated RIP3 (Receptor-Interacting Protein Kinase 3) and NLRP3 (NOD-Like Receptor Pyrin Domain Containing 3) Inflammasome Activation. Circulation 2020; 141:984-1000. [PMID: 31902237 DOI: 10.1161/circulationaha.119.042336] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND S-nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in the pathogenesis of cardiovascular disease. The aim of this study was to determine the role of SNO of MLP (muscle LIM protein) in myocardial hypertrophy, as well as the mechanism by which SNO-MLP modulates hypertrophic growth in response to pressure overload. METHODS Myocardial samples from patients and animal models exhibiting myocardial hypertrophy were examined for SNO-MLP level using biotin-switch methods. SNO sites were further identified through liquid chromatography-tandem mass spectrometry. Denitrosylation of MLP by the mutation of nitrosylation sites or overexpression of S-nitrosoglutathione reductase was used to analyze the contribution of SNO-MLP in myocardial hypertrophy. Downstream effectors of SNO-MLP were screened through mass spectrometry and confirmed by coimmunoprecipitation. Recruitment of TLR3 (Toll-like receptor 3) by SNO-MLP in myocardial hypertrophy was examined in TLR3 small interfering RNA-transfected neonatal rat cardiomyocytes and in a TLR3 knockout mouse model. RESULTS SNO-MLP level was significantly higher in hypertrophic myocardium from patients and in spontaneously hypertensive rats and mice subjected to transverse aortic constriction. The level of SNO-MLP also increased in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes. S-nitrosylated site of MLP at cysteine 79 was identified by liquid chromatography-tandem mass spectrometry and confirmed in neonatal rat cardiomyocytes. Mutation of cysteine 79 significantly reduced hypertrophic growth in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes and transverse aortic constriction mice. Reducing SNO-MLP level by overexpression of S-nitrosoglutathione reductase greatly attenuated myocardial hypertrophy. Mechanistically, SNO-MLP stimulated TLR3 binding to MLP in response to hypertrophic stimuli, and disrupted this interaction by downregulating TLR3-attenuated myocardial hypertrophy. SNO-MLP also increased the complex formation between TLR3 and RIP3 (receptor-interacting protein kinase 3). This interaction in turn induced NLRP3 (nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3) inflammasome activation, thereby promoting the development of myocardial hypertrophy. CONCLUSIONS Our findings revealed a key role of SNO-MLP in myocardial hypertrophy and demonstrated TLR3-mediated RIP3 and NLRP3 inflammasome activation as the downstream signaling pathway, which may represent a therapeutic target for myocardial hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Lihong Pan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Feiyue Dai
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Menglin Chao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Hong Jiang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Xuesong Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhe Lin
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhengrong Huang
- Department of Cardiology, the First Affiliated Hospital of Xiamen University, China (Z.H.)
| | - Guoliang Meng
- Nanjing Medical University, Nanjing, China (G.M.).,Department of Pharmacology, School of Pharmacy, Nantong University, China (G.M.)
| | - Chun Wang
- Department of Geriatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, China (C.W.)
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Xin Wang
- Faculty of Biology, Medicine and Health, the University of Manchester, United Kingdom (X.W.)
| | - Albert Ferro
- Cardiovascular Clinical Pharmacology, British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, United Kingdom (A.F.)
| | - Hong Wang
- Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA (H.W.)
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yi Han
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, China (Y.H.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.).,State Key Laboratory of Reproductive Medicine (Y.J.)
| |
Collapse
|
84
|
Newly Discovered Micropeptide Regulators of SERCA Form Oligomers but Bind to the Pump as Monomers. J Mol Biol 2019; 431:4429-4443. [PMID: 31449798 DOI: 10.1016/j.jmb.2019.07.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
The recently-discovered single-span transmembrane proteins endoregulin (ELN), dwarf open reading frame (DWORF), myoregulin (MLN), and another-regulin (ALN) are reported to bind to the SERCA calcium pump in a manner similar to that of known regulators of SERCA activity, phospholamban (PLB) and sarcolipin (SLN). To determine how micropeptide assembly into oligomers affects the availability of the micropeptide to bind to SERCA in a regulatory complex, we used co-immunoprecipitation and fluorescence resonance energy transfer (FRET) to quantify micropeptide oligomerization and SERCA-binding. Micropeptides formed avid homo-oligomers with high-order stoichiometry (n > 2 protomers per homo-oligomer), but it was the monomeric form of all micropeptides that interacted with SERCA. In view of these two alternative binding interactions, we evaluated the possibility that oligomerization occurs at the expense of SERCA-binding. However, even the most avidly oligomeric micropeptide species still showed robust FRET with SERCA, and there was a surprising positive correlation between oligomerization affinity and SERCA-binding. This comparison of micropeptide family members suggests that the same structural determinants that support oligomerization are also important for binding to SERCA. Moreover, the unique oligomerization/SERCA-binding profile of DWORF is in harmony with its distinct role as a PLB-competing SERCA activator, in contrast to the inhibitory function of the other SERCA-binding micropeptides.
Collapse
|
85
|
Abstract
PURPOSE OF REVIEW Human genome is pervasively transcribed, producing coding and noncoding RNAs. Recent studies have revealed the roles of a class of noncoding RNAs, the long noncoding RNAs (lncRNAs), in heart failure and other cardiovascular diseases. This review provides a brief summary of recent findings on lncRNA function. RECENT FINDINGS Recent studies have documented the roles of lncRNAs in cardiac regeneration, conduction, hypertrophy/dysfunction, and endothelial function. LncRNAs perform these functions through acting as competing RNA (by binding and sequestering MicroRNAs) or acting as guides to protein targeting. A few lncRNAs also encode small peptides (e.g., Dwarf Open Reading Frame RNA) and in the context of heart regulate cardiac calcium homeostasis. SUMMARY Noncoding RNA provides a versatile mechanism of gene regulation and thereby present as novel targets for intervention in various cardiovascular disease. Future studies aimed at defining the context-dependent lncRNA mechanisms will be required to advance our understanding and relish the goal of RNA therapeutics.
Collapse
Affiliation(s)
- Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, and Texas Heart Institute, Houston, TX 77030., Tel: +1 713-500-2335,
| |
Collapse
|
86
|
Kerkhofs M, Bultynck G, Vervliet T, Monaco G. Therapeutic implications of novel peptides targeting ER-mitochondria Ca 2+-flux systems. Drug Discov Today 2019; 24:1092-1103. [PMID: 30910738 DOI: 10.1016/j.drudis.2019.03.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/16/2019] [Accepted: 03/18/2019] [Indexed: 01/03/2023]
Abstract
Intracellular Ca2+-flux systems located at the ER-mitochondrial axis govern mitochondrial Ca2+ balance and cell fate. Multiple yet incurable pathologies are characterized by insufficient or excessive Ca2+ fluxes toward the mitochondria, in turn leading to aberrant cell life or death dynamics. The discovery and ongoing molecular characterization of the main interorganellar Ca2+ gateways have resulted in a novel class of peptide tools able to regulate relevant protein-protein interactions (PPIs) underlying this signaling scenario. Here, we review peptides, molecularly derived from Ca2+-flux systems or their accessory proteins. We discuss how they alter Ca2+-signaling protein complexes and modulate cell survival in light of their forthcoming therapeutic applications.
Collapse
Affiliation(s)
- Martijn Kerkhofs
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium.
| | - Tim Vervliet
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Giovanni Monaco
- KU Leuven, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Laboratory of Molecular and Cellular Signaling, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
87
|
Khitun A, Ness TJ, Slavoff SA. Small open reading frames and cellular stress responses. Mol Omics 2019; 15:108-116. [PMID: 30810554 DOI: 10.1039/c8mo00283e] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small open reading frames (smORFs) encoding polypeptides of less than 100 amino acids in eukaryotes (50 amino acids in prokaryotes) were historically excluded from genome annotation. However, recent advances in genomics, ribosome footprinting, and proteomics have revealed thousands of translated smORFs in genomes spanning evolutionary space. These smORFs can encode functional polypeptides, or act as cis-translational regulators. Herein we review evidence that some smORF-encoded polypeptides (SEPs) participate in stress responses in both prokaryotes and eukaryotes, and that some upstream ORFs (uORFs) regulate stress-responsive translation of downstream cistrons in eukaryotic cells. These studies provide insight into a regulated subclass of smORFs and suggest that at least some SEPs may participate in maintenance of cellular homeostasis under stress.
Collapse
Affiliation(s)
- Alexandra Khitun
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Travis J Ness
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Sarah A Slavoff
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA and Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| |
Collapse
|