1
|
Viskupicova J, Michel Espinoza-Fonseca L. Allosteric modulation of SERCA pumps in health and disease: structural dynamics, posttranslational modifications, and therapeutic potential. J Mol Biol 2025:169200. [PMID: 40349954 DOI: 10.1016/j.jmb.2025.169200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Sarco/endoplasmic reticulum (SR/ER) Ca2+-ATPase (SERCA) pumps are ubiquitous membrane proteins in all eukaryotic cells, playing a central role in maintaining intracellular calcium homeostasis by re-sequestering Ca2+ ions from the cytosol into the SR/ER at the expense of ATP hydrolysis. SERCA pumps are well-characterized components of the calcium transport machinery in the cell, playing a role in various physiological processes, including muscle contraction, energy metabolism, secretion exocytosis, gene expression, synaptic transmission, cell survival, and fertilization. Allosteric regulation of SERCA pumps plays a key role in health and disease, and modulation of the SERCA pumps has emerged as a therapeutic approach for the treatment of cardiovascular, muscular, metabolic, and neurodegenerative disorders. In this review, we provide a comprehensive overview of the structural dynamics underlying allosteric modulation of SERCA, focusing on the effects of endogenous regulatory proteins, Ca2+ ions, ATP, and small-molecule effectors on the dynamics and function of the pump. We also examine in detail the role of posttranslational modifications as allosteric modulators of SERCA function, focusing on the oxidative modifications S-glutathionylation, S-nitrosylation, tyrosine nitration, and carbonylation, and non-oxidative modifications that include SUMOylation, acetylation, O-GlcNAcylation, phosphorylation, and ubiquitination. Finally, we discuss the therapeutic potential and challenges of allosteric modulation of SERCA pumps, including the design of small-molecule effectors, microRNA-based interventions, and targeted strategies that modulate SERCA posttranslational regulation. Overall, this review aims to bridge the gap between the mechanisms underlying allosteric modulation of SERCA and the translation of basic science discoveries into effective therapies targeting SERCA pumps.
Collapse
Affiliation(s)
- Jana Viskupicova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology & Toxicology, Slovak Academy of Sciences, 84104 Bratislava, Slovakia.
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Weber DK, Reddy UV, Robia SL, Veglia G. Pathological mutations in the phospholamban cytoplasmic region affect its topology and dynamics modulating the extent of SERCA inhibition. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184370. [PMID: 38986894 PMCID: PMC11457527 DOI: 10.1016/j.bbamem.2024.184370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Phospholamban (PLN) is a 52 amino acid regulin that allosterically modulates the activity of the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) in the heart muscle. In its unphosphorylated form, PLN binds SERCA within its transmembrane (TM) domains, approximately 20 Å away from the Ca2+ binding site, reducing SERCA's apparent Ca2+ affinity (pKCa) and decreasing cardiac contractility. During the enzymatic cycle, the inhibitory TM domain of PLN remains anchored to SERCA, whereas its cytoplasmic region transiently binds the ATPase's headpiece. Phosphorylation of PLN at Ser16 by protein kinase A increases the affinity of its cytoplasmic domain to SERCA, weakening the TM interactions with the ATPase, reversing its inhibitory function, and augmenting muscle contractility. How the structural changes caused by pathological mutations in the PLN cytoplasmic region are transmitted to its inhibitory TM domain is still unclear. Using solid-state NMR spectroscopy and activity assays, we analyzed the structural and functional effects of a series of mutations and their phosphorylated forms located in the PLN cytoplasmic region and linked to dilated cardiomyopathy. We found that these missense mutations affect the overall topology and dynamics of PLN and ultimately modulate its inhibitory potency. Also, the changes in the TM tilt angle and cytoplasmic dynamics of PLN caused by these mutations correlate well with the extent of SERCA inhibition. Our study unveils new molecular determinants for designing variants of PLN that outcompete endogenous PLN to regulate SERCA in a tunable manner.
Collapse
Affiliation(s)
- Daniel K Weber
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - U Venkateswara Reddy
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Dowrick JM, Tran K, Garrett AS, Anderson AJ, Nielsen PMF, Taberner AJ, Han JC. Work-loop contractions reveal that the afterload-dependent time course of cardiac Ca 2+ transients is modulated by preload. J Appl Physiol (1985) 2022; 133:663-675. [PMID: 35771221 PMCID: PMC9762964 DOI: 10.1152/japplphysiol.00137.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Preload and afterload dictate the dynamics of the cyclical work-loop contraction that the heart undergoes in vivo. Cellular Ca2+ dynamics drive contraction, but the effects of afterload alone on the Ca2+ transient are inconclusive. To our knowledge, no study has investigated whether the putative afterload dependence of the Ca2+ transient is preload dependent. This study is designed to provide the first insight into the Ca2+ handling of cardiac trabeculae undergoing work-loop contractions, with the aim to examine whether the conflicting afterload dependency of the Ca2+ transient can be accounted for by considering preload under isometric and physiological work-loop contractions. Thus, we subjected ex vivo rat right-ventricular trabeculae, loaded with the fluorescent dye Fura-2, to work-loop contractions over a wide range of afterloads at two preloads while measuring stress, length changes, and Ca2+ transients. Work-loop control was implemented with a real-time Windkessel model to mimic the contraction patterns of the heart in vivo. We extracted a range of metrics from the measured steady-state twitch stress and Ca2+ transients, including the amplitudes, time courses, rates of rise, and integrals. Results show that parameters of stress were afterload and preload dependent. In contrast, the parameters associated with Ca2+ transients displayed a mixed dependence on afterload and preload. Most notably, its time course was afterload dependent, an effect augmented at the greater preload. This study reveals that the afterload dependence of cardiac Ca2+ transients is modulated by preload, which brings the study of Ca2+ transients during isometric contractions into question when aiming to understand physiological Ca2+ handling.NEW & NOTEWORTHY This study is the first examination of Ca2+ handling in trabeculae undergoing work-loop contractions. These data reveal that reducing preload diminishes the influence of afterload on the decay phase of the cardiac Ca2+ transient. This is significant as it reconciles inconsistencies in the literature regarding the influence of external loads on cardiac Ca2+ handling. Furthermore, these findings highlight discrepancies between Ca2+ handling during isometric and work-loop contractions in cardiac trabeculae operating at their optimal length.
Collapse
Affiliation(s)
- Jarrah M. Dowrick
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Kenneth Tran
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Amy S. Garrett
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alex J. Anderson
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Poul M. F. Nielsen
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand,2Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - Andrew J. Taberner
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand,2Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - June-Chiew Han
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Benkner A, Rüdebusch J, Nath N, Hammer E, Grube K, Gross S, Dhople VM, Eckstein G, Meitinger T, Kaderali L, Völker U, Fielitz J, Felix SB. Riociguat attenuates left ventricular proteome and microRNA profile changes after experimental aortic stenosis in mice. Br J Pharmacol 2022; 179:4575-4592. [PMID: 35751875 DOI: 10.1111/bph.15910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 04/15/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Development and progression of heart failure (HF) involve endothelial and myocardial dysfunction as well as a dysregulation of the nitric oxide - soluble guanylyl cyclase - cyclic guanosine monophosphate (NO-sGC-cGMP) signalling pathway. Recently, we reported that the sGC stimulator riociguat (RIO) has beneficial effects on cardiac remodelling and progression of HF in response to chronic pressure overload. Here, we examined if these favourable RIO effects are also reflected in alterations of the myocardial proteome and microRNA profiles. EXPERIMENTAL APPROACH Male C57BL/6N mice underwent transverse aortic constriction (TAC) and sham operated mice served as controls. TAC and sham animals were randomised and treated with either RIO or vehicle for five weeks, starting three weeks post-surgery when cardiac hypertrophy was established. Afterwards we performed mass spectrometric proteome analyses and microRNA sequencing of proteins and RNAs, respectively, isolated from left ventricles (LV). KEY RESULTS TAC-induced changes of the LV proteome were significantly reduced by RIO treatment. Bioinformatics analyses revealed that RIO improved TAC-induced cardiovascular disease related pathways, metabolism and energy production, e.g. reversed alterations in the levels of myosin heavy chain 7 (MYH7), cardiac phospholamban (PLN), and ankyrin repeat domain-containing protein 1 (ANKRD1). RIO also attenuated TAC-induced changes of microRNA levels in the LV. CONCLUSION AND IMPLICATIONS The sGC stimulator RIO has beneficial effects on cardiac structure and function during pressure overload, which is accompanied by a reversal of TAC-induced changes of the cardiac proteome and microRNA profile. Our data support the potential of RIO as a novel HF therapeutic.
Collapse
Affiliation(s)
- Alexander Benkner
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Julia Rüdebusch
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Neetika Nath
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Karina Grube
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Gross
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Vishnu M Dhople
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Gertrud Eckstein
- Institute of Human Genetics, Helmholtz Centre Munich, Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Centre Munich, Neuherberg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Lars Kaderali
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jens Fielitz
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B Felix
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
CRISPR/Cas9-mediated tryptophan hydroxylase 1 knockout decreases calcium transportation in goat mammary epithelial cells. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
6
|
Non-Coding RNAs in the Cardiac Action Potential and Their Impact on Arrhythmogenic Cardiac Diseases. HEARTS 2021. [DOI: 10.3390/hearts2030026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cardiac arrhythmias are prevalent among humans across all age ranges, affecting millions of people worldwide. While cardiac arrhythmias vary widely in their clinical presentation, they possess shared complex electrophysiologic properties at cellular level that have not been fully studied. Over the last decade, our current understanding of the functional roles of non-coding RNAs have progressively increased. microRNAs represent the most studied type of small ncRNAs and it has been demonstrated that miRNAs play essential roles in multiple biological contexts, including normal development and diseases. In this review, we provide a comprehensive analysis of the functional contribution of non-coding RNAs, primarily microRNAs, to the normal configuration of the cardiac action potential, as well as their association to distinct types of arrhythmogenic cardiac diseases.
Collapse
|
7
|
Karanja CW, Yeboah KS, Ong WWS, Sintim HO. A STING-based fluorescent polarization assay for monitoring activities of cyclic dinucleotide metabolizing enzymes. RSC Chem Biol 2021; 2:206-214. [PMID: 34458783 PMCID: PMC8341399 DOI: 10.1039/d0cb00187b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022] Open
Abstract
Cyclic dinucleoties, such as cGAMP, c-di-GMP and c-di-AMP, are fascinating second messengers with diverse roles in both prokaryotes and eukaryotes. Consequently there is a need for simple and inexpensive methods for profiling these compounds in biological media, monitoring their synthesis or degradation by enzymes and for identifying inhibitors of proteins that metabolize or bind to these dinucleotides. Since 2011, when we reported the first simple method to detect c-di-GMP (S. Nakayama, I. Kelsey, J. Wang, K. Roelofs, B. Stefane, Y. Luo, V. T. Lee and H. O. Sintim, J. Am. Chem. Soc., 2011, 133, 4856) or in 2014 when we revealed another surprisingly simple assay to detect c-di-AMP (J. Zhou, D. A. Sayre, Y. Zheng, H. Szmacinski and H. O. Sintim, Anal. Chem., 2014, 86, 2412), there have been efforts to develop assays to detect cyclic dinucleotides by others. However a unified and simple assay, which can be used for all cyclic dinucleotides is lacking. Here, we investigate STING binding by various fluorescein-labeled c-di-GMP, c-di-AMP and cGAMP, using fluorescent polarization (FP). Fluorescein-labeled c-di-GMP (F-c-di-GMP) was found to be the best binder of STING. This probe could be displaced by unlabeled cGAMP, c-di-AMP or c-di-GMP and hence it is a universal probe, which can be used to monitor all three dinucleotides. HPLC analysis was used to validate the new F-c-di-GMP-based FP assay.
Collapse
Affiliation(s)
- Caroline W Karanja
- Department of Chemistry 560 Oval Drive West Lafayette Indiana 47907-2084 USA
| | - Kofi S Yeboah
- Department of Chemistry 560 Oval Drive West Lafayette Indiana 47907-2084 USA
| | - Wilson W S Ong
- Department of Chemistry 560 Oval Drive West Lafayette Indiana 47907-2084 USA
| | - Herman O Sintim
- Department of Chemistry 560 Oval Drive West Lafayette Indiana 47907-2084 USA
- Institute for Drug Discovery, Purdue University 720 Clinic Drive West Lafayette IN 47907 USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease West Lafayette IN 47907 USA
| |
Collapse
|
8
|
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
|
9
|
Stroik DR, Ceholski DK, Bidwell PA, Mleczko J, Thanel PF, Kamdar F, Autry JM, Cornea RL, Thomas DD. Viral expression of a SERCA2a-activating PLB mutant improves calcium cycling and synchronicity in dilated cardiomyopathic hiPSC-CMs. J Mol Cell Cardiol 2019; 138:59-65. [PMID: 31751570 DOI: 10.1016/j.yjmcc.2019.11.147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Abstract
There is increasing momentum toward the development of gene therapy for heart failure (HF) that is defined by impaired calcium (Ca2+) transport and reduced contractility. We have used FRET (fluorescence resonance energy transfer) between fluorescently-tagged SERCA2a (the cardiac Ca2+ pump) and PLB (phospholamban, ventricular peptide inhibitor of SERCA) to test directly the effectiveness of loss-of-inhibition/gain-of-binding (LOI/GOB) PLB mutants (PLBM) that were engineered to compete with the binding of inhibitory wild-type PLB (PLBWT). Our therapeutic strategy is to relieve PLBWT inhibition of SERCA2a by using the reserve adrenergic capacity mediated by PLB to enhance cardiac contractility. Using a FRET assay, we determined that the combination of a LOI PLB mutation (L31A) and a GOB PLB mutation (I40A) results in a novel engineered LOI/GOB PLBM (L31A/I40A) that effectively competes with PLBWT binding to cardiac SERCA2a in HEK293-6E cells. We demonstrated that co-expression of PLBM enhances SERCA Ca-ATPase activity by increasing enzyme Ca2+ affinity (1/KCa) in PLBWT-inhibited HEK293 cell homogenates. For an initial assessment of PLBM physiological effectiveness, we used human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) from a healthy individual. In this system, we observed that adeno-associated virus 2 (rAAV2)-driven expression of PLBM enhances the amplitude of SR Ca2+ release and the rate of SR Ca2+ re-uptake. To assess therapeutic potential, we used a hiPSC-CM model of dilated cardiomyopathy (DCM) containing PLB mutation R14del, where we observed that rAAV2-driven expression of PLBM rescues arrhythmic Ca2+ transients and alleviates decreased Ca2+ transport. Thus, we propose that PLBM transgene expression is a promising gene therapy strategy that directly targets the underlying pathophysiology of abnormal Ca2+ transport and thus contractility in underlying systolic heart failure.
Collapse
Affiliation(s)
- Daniel R Stroik
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Delaine K Ceholski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Philip A Bidwell
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Justyna Mleczko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Paul F Thanel
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Forum Kamdar
- Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Joseph M Autry
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
10
|
Mayourian J, Ceholski DK, Gonzalez DM, Cashman TJ, Sahoo S, Hajjar RJ, Costa KD. Physiologic, Pathologic, and Therapeutic Paracrine Modulation of Cardiac Excitation-Contraction Coupling. Circ Res 2019; 122:167-183. [PMID: 29301848 DOI: 10.1161/circresaha.117.311589] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac excitation-contraction coupling (ECC) is the orchestrated process of initial myocyte electrical excitation, which leads to calcium entry, intracellular trafficking, and subsequent sarcomere shortening and myofibrillar contraction. Neurohumoral β-adrenergic signaling is a well-established mediator of ECC; other signaling mechanisms, such as paracrine signaling, have also demonstrated significant impact on ECC but are less well understood. For example, resident heart endothelial cells are well-known physiological paracrine modulators of cardiac myocyte ECC mainly via NO and endothelin-1. Moreover, recent studies have demonstrated other resident noncardiomyocyte heart cells (eg, physiological fibroblasts and pathological myofibroblasts), and even experimental cardiotherapeutic cells (eg, mesenchymal stem cells) are also capable of altering cardiomyocyte ECC through paracrine mechanisms. In this review, we first focus on the paracrine-mediated effects of resident and therapeutic noncardiomyocytes on cardiomyocyte hypertrophy, electrophysiology, and calcium handling, each of which can modulate ECC, and then discuss the current knowledge about key paracrine factors and their underlying mechanisms of action. Next, we provide a case example demonstrating the promise of tissue-engineering approaches to study paracrine effects on tissue-level contractility. More specifically, we present new functional and molecular data on the effects of human adult cardiac fibroblast conditioned media on human engineered cardiac tissue contractility and ion channel gene expression that generally agrees with previous murine studies but also suggests possible species-specific differences. By contrast, paracrine secretions by human dermal fibroblasts had no discernible effect on human engineered cardiac tissue contractile function and gene expression. Finally, we discuss systems biology approaches to help identify key stem cell paracrine mediators of ECC and their associated mechanistic pathways. Such integration of tissue-engineering and systems biology methods shows promise to reveal novel insights into paracrine mediators of ECC and their underlying mechanisms of action, ultimately leading to improved cell-based therapies for patients with heart disease.
Collapse
Affiliation(s)
- Joshua Mayourian
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Delaine K Ceholski
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David M Gonzalez
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Timothy J Cashman
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Susmita Sahoo
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
11
|
Thompson BR, Soller KJ, Vetter A, Yang J, Veglia G, Bowser MT, Metzger JM. Cytoplasmic nucleic acid-based XNAs directly enhance live cardiac cell function by a Ca 2+ cycling-independent mechanism via the sarcomere. J Mol Cell Cardiol 2019; 130:1-9. [PMID: 30849419 DOI: 10.1016/j.yjmcc.2019.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/05/2019] [Accepted: 02/27/2019] [Indexed: 11/19/2022]
Abstract
Nucleic acid - protein interactions are critical for regulating gene activation in the nucleus. In the cytoplasm, however, potential nucleic acid-protein functional interactions are less clear. The emergence of a large and expanding number of non-coding RNAs and DNA fragments raises the possibility that the cytoplasmic nucleic acids may interact with cytoplasmic cellular components to directly alter key biological processes within the cell. We now show that both natural and synthetic nucleic acids, collectively XNAs, when introduced to the cytoplasm of live cell cardiac myocytes, markedly enhance contractile function via a mechanism that is independent of new translation, activation of the TLR-9 pathway or by altered intracellular Ca2+ cycling. Findings show a steep XNA oligo length-dependence, but not sequence dependence or nucleic acid moiety dependence, for cytoplasmic XNAs to hasten myocyte relaxation. XNAs localized to the sarcomere in a striated pattern and bound the cardiac troponin regulatory complex with high affinity in an electrostatic-dependent manner. Mechanistically, XNAs phenocopy PKA-based modified troponin to cause faster relaxation. Collectively, these data support a new role for cytoplasmic nucleic acids in directly modulating live cell cardiac performance and raise the possibility that cytoplasmic nucleic acid - protein interactions may alter functionally relevant pathways in other cell types.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Kailey J Soller
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Anthony Vetter
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Jing Yang
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Michael T Bowser
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America.
| |
Collapse
|
12
|
Sahu B, Pani S, Swalsingh G, Bal NC. Non and Epigenetic Mechanisms in Regulation of Adaptive Thermogenesis in Skeletal Muscle. Front Endocrinol (Lausanne) 2019; 10:517. [PMID: 31456746 PMCID: PMC6700214 DOI: 10.3389/fendo.2019.00517] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/15/2019] [Indexed: 01/07/2023] Open
|
13
|
Adão R, Mendes-Ferreira P, Maia-Rocha C, Santos-Ribeiro D, Rodrigues PG, Vidal-Meireles A, Monteiro-Pinto C, Pimentel LD, Falcão-Pires I, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Neuregulin-1 attenuates right ventricular diastolic stiffness in experimental pulmonary hypertension. Clin Exp Pharmacol Physiol 2018; 46:255-265. [PMID: 30339273 DOI: 10.1111/1440-1681.13043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 12/28/2022]
Abstract
We have previously shown that treatment with recombinant human neuregulin-1 (rhNRG-1) improves pulmonary arterial hypertension (PAH) in a monocrotaline (MCT)-induced animal model, by decreasing pulmonary arterial remodelling and endothelial dysfunction, as well as by restoring right ventricular (RV) function. Additionally, rhNRG-1 treatment showed direct myocardial anti-remodelling effects in a model of pressure loading of the RV without PAH. This work aimed to study the intrinsic cardiac effects of rhNRG-1 on experimental PAH and RV pressure overload, and more specifically on diastolic stiffness, at both the ventricular and cardiomyocyte level. We studied the effects of chronic rhNRG-1 treatment on ventricular passive stiffness in RV and LV samples from MCT-induced PAH animals and in the RV from animals with compensated and decompensated RV hypertrophy, through a mild and severe pulmonary artery banding (PAB). We also measured passive tension in isolated cardiomyocytes and quantified the expression of myocardial remodelling-associated genes and calcium handling proteins. Chronic rhNRG-1 treatment decreased passive tension development in RV and LV isolated from animals with MCT-induced PAH. This decrease was associated with increased phospholamban phosphorylation, and with attenuation of the expression of cardiac maladaptive remodelling markers. Finally, we showed that rhNRG-1 therapy decreased RV remodelling and cardiomyocyte passive tension development in PAB-induced RV hypertrophy animals, without compromising cardiac function, pointing to cardiac-specific effects in both hypertrophy stages. In conclusion, we demonstrated that rhNRG-1 treatment decreased RV intrinsic diastolic stiffness, through the improvement of calcium handling and cardiac remodelling signalling.
Collapse
Affiliation(s)
- Rui Adão
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Pedro Mendes-Ferreira
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Carolina Maia-Rocha
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diana Santos-Ribeiro
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Patrícia Gonçalves Rodrigues
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - André Vidal-Meireles
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Cláudia Monteiro-Pinto
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Luís D Pimentel
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, UnIC-Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal.,Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
14
|
Mayourian J, Ceholski DK, Gorski PA, Mathiyalagan P, Murphy JF, Salazar SI, Stillitano F, Hare JM, Sahoo S, Hajjar RJ, Costa KD. Exosomal microRNA-21-5p Mediates Mesenchymal Stem Cell Paracrine Effects on Human Cardiac Tissue Contractility. Circ Res 2018; 122:933-944. [PMID: 29449318 DOI: 10.1161/circresaha.118.312420] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 01/08/2023]
Abstract
RATIONALE The promising clinical benefits of delivering human mesenchymal stem cells (hMSCs) for treating heart disease warrant a better understanding of underlying mechanisms of action. hMSC exosomes increase myocardial contractility; however, the exosomal cargo responsible for these effects remains unresolved. OBJECTIVE This study aims to identify lead cardioactive hMSC exosomal microRNAs to provide a mechanistic basis for optimizing future stem cell-based cardiotherapies. METHODS AND RESULTS Integrating systems biology and human engineered cardiac tissue (hECT) technologies, partial least squares regression analysis of exosomal microRNA profiling data predicted microRNA-21-5p (miR-21-5p) levels positively correlate with contractile force and calcium handling gene expression responses in hECTs treated with conditioned media from multiple cell types. Furthermore, miR-21-5p levels were significantly elevated in hECTs treated with the exosome-enriched fraction of the hMSC secretome (hMSC-exo) versus untreated controls. This motivated experimentally testing the human-specific role of miR-21-5p in hMSC-exo-mediated increases of cardiac tissue contractility. Treating hECTs with miR-21-5p alone was sufficient to recapitulate effects observed with hMSC-exo on hECT developed force and expression of associated calcium handling genes (eg, SERCA2a and L-type calcium channel). Conversely, knockdown of miR-21-5p in hMSCs significantly diminished exosomal procontractile and associated calcium handling gene expression effects on hECTs. Western blots supported miR-21-5p effects on calcium handling gene expression at the protein level, corresponding to significantly increased calcium transient amplitude and decreased decay time constant in comparison to miR-scramble control. Mechanistically, cotreating with miR-21-5p and LY294002, a PI3K inhibitor, suppressed these effects. Finally, mathematical simulations predicted the translational capacity for miR-21-5p treatment to restore calcium handling in mature ischemic adult human cardiomyocytes. CONCLUSIONS miR-21-5p plays a key role in hMSC-exo-mediated effects on cardiac contractility and calcium handling, likely via PI3K signaling. These findings may open new avenues of research to harness the role of miR-21-5p in optimizing future stem cell-based cardiotherapies.
Collapse
Affiliation(s)
- Joshua Mayourian
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Delaine K Ceholski
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Przemek A Gorski
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Prabhu Mathiyalagan
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Jack F Murphy
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Sophia I Salazar
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Francesca Stillitano
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Joshua M Hare
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Susmita Sahoo
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.)
| | - Kevin D Costa
- From the Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (J.M., D.K.C., P.A.G., P.M., J.F.M., S.I.S., F.S., S.S., R.J.H., K.D.C.); and Interdisciplinary Stem Cell Institute, Department of Cardiology, University of Miami Miller School of Medicine, Miami, FL (J.M.H.).
| |
Collapse
|
15
|
Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR, Ge ZD. Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep 2017; 7:3093. [PMID: 28596578 PMCID: PMC5465102 DOI: 10.1038/s41598-017-03234-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
GTP cyclohydrolase 1 (GCH1) and its product tetrahydrobiopterin play crucial roles in cardiovascular health and disease, yet the exact regulation and role of GCH1 in adverse cardiac remodeling after myocardial infarction are still enigmatic. Here we report that cardiac GCH1 is degraded in remodeled hearts after myocardial infarction, concomitant with increases in the thickness of interventricular septum, interstitial fibrosis, and phosphorylated p38 mitogen-activated protein kinase and decreases in left ventricular anterior wall thickness, cardiac contractility, tetrahydrobiopterin, the dimers of nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and the expression of sarcoplasmic reticulum Ca2+ handling proteins. Intriguingly, transgenic overexpression of GCH1 in cardiomyocytes reduces the thickness of interventricular septum and interstitial fibrosis and increases anterior wall thickness and cardiac contractility after infarction. Moreover, we show that GCH1 overexpression decreases phosphorylated p38 mitogen-activated protein kinase and elevates tetrahydrobiopterin levels, the dimerization and phosphorylation of neuronal nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and sarcoplasmic reticulum Ca2+ handling proteins in post-infarction remodeled hearts. Our results indicate that the pivotal role of GCH1 overexpression in post-infarction cardiac remodeling is attributable to preservation of neuronal nitric oxide synthase and sarcoplasmic reticulum Ca2+ handling proteins, and identify a new therapeutic target for cardiac remodeling after infarction.
Collapse
Affiliation(s)
- Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Medicine, Columbia University, 630 W. 168th Street, New York, New York, 10032, USA
| | - Shelley L Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, Wisconsin, 53234, USA
| | - Shigang Qiao
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zeljko J Bosnjak
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Physiology, Medical College of Wiscosin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, People's Republic of China
| | - David C Warltier
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Judy R Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.
| |
Collapse
|
16
|
张 建, 吴 进, 李 士, 龚 园. [Lowered sarcoendoplasmic reticulum calcium uptake and diaphragmatic SERCA1 expression contribute to diaphragmatic contractile and relaxation dysfunction in septic rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2016; 37:438-443. [PMID: 28446393 PMCID: PMC6744105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Indexed: 10/15/2023]
Abstract
OBJECTIVE The explore the mechanism responsible for diaphragmatic contractile and relaxation dysfunction in a rat model of sepsis. METHODS Thirty-six adult male Sprague-Dawley rats were randomized equally into a sham-operated group and two model groups of sepsis induced by cecal ligation and puncture (CLP) for examination at 6 and 12 h following CLP (CLP-6 h and CLP-12 h groups). The parameters of diaphragm contractile and relaxation were measured, and the calcium uptake and release rates of the diaphragmatic sarcoendoplasmic reticulum (SR) and the protein expressions of SERCA1, SERCA2 and RyR in the diaphragmatic muscles were determined. RESULTS The half-relaxation time of the diaphragm was extended in both the CLP-6 h and CLP-12 h groups with significantly reduced maximum tension declinerate and the peek uptake rate of SERCA (P<0.01). Diaphragmatic maximum twitch force development rate, the maximal twitch, tetanus tensions and the peek release rate of SR decreased only at 12h after CLP (P<0.01). The expression levels of SERCA1 protein decreased significantly in the diaphragmatic muscles at 12h following CLP (P<0.01) while SERCA2 expression level and SERCA activity showed no significant changes. CONCLUSION In the acute stage of sepsis, both the contractile and relaxation functions of the diaphragm are impaired. Diaphragmatic relaxation dysfunction may result from reduced calcium uptake in the SR and a decreased level of SERCA1 in the diaphragmatic muscles.
Collapse
Affiliation(s)
- 建友 张
- 南京医科大学,江苏 南京 210029Nanjing Medical University, Nanjing 210029, China
| | - 进 吴
- 上海交通大学附属第一人民医院麻醉科,上海 200080Department of Anesthesiology, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, China
| | - 士通 李
- 南京医科大学,江苏 南京 210029Nanjing Medical University, Nanjing 210029, China
| | - 园 龚
- 上海交通大学附属第一人民医院麻醉科,上海 200080Department of Anesthesiology, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, China
| |
Collapse
|
17
|
张 建, 吴 进, 李 士, 龚 园. [Lowered sarcoendoplasmic reticulum calcium uptake and diaphragmatic SERCA1 expression contribute to diaphragmatic contractile and relaxation dysfunction in septic rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2016; 37:438-443. [PMID: 28446393 PMCID: PMC6744105 DOI: 10.3969/j.issn.1673-4254.2017.04.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE The explore the mechanism responsible for diaphragmatic contractile and relaxation dysfunction in a rat model of sepsis. METHODS Thirty-six adult male Sprague-Dawley rats were randomized equally into a sham-operated group and two model groups of sepsis induced by cecal ligation and puncture (CLP) for examination at 6 and 12 h following CLP (CLP-6 h and CLP-12 h groups). The parameters of diaphragm contractile and relaxation were measured, and the calcium uptake and release rates of the diaphragmatic sarcoendoplasmic reticulum (SR) and the protein expressions of SERCA1, SERCA2 and RyR in the diaphragmatic muscles were determined. RESULTS The half-relaxation time of the diaphragm was extended in both the CLP-6 h and CLP-12 h groups with significantly reduced maximum tension declinerate and the peek uptake rate of SERCA (P<0.01). Diaphragmatic maximum twitch force development rate, the maximal twitch, tetanus tensions and the peek release rate of SR decreased only at 12h after CLP (P<0.01). The expression levels of SERCA1 protein decreased significantly in the diaphragmatic muscles at 12h following CLP (P<0.01) while SERCA2 expression level and SERCA activity showed no significant changes. CONCLUSION In the acute stage of sepsis, both the contractile and relaxation functions of the diaphragm are impaired. Diaphragmatic relaxation dysfunction may result from reduced calcium uptake in the SR and a decreased level of SERCA1 in the diaphragmatic muscles.
Collapse
Affiliation(s)
- 建友 张
- 南京医科大学,江苏 南京 210029Nanjing Medical University, Nanjing 210029, China
| | - 进 吴
- 上海交通大学附属第一人民医院麻醉科,上海 200080Department of Anesthesiology, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, China
| | - 士通 李
- 南京医科大学,江苏 南京 210029Nanjing Medical University, Nanjing 210029, China
| | - 园 龚
- 上海交通大学附属第一人民医院麻醉科,上海 200080Department of Anesthesiology, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, China
| |
Collapse
|