1
|
Zhao WB, Sheng R. The correlation between mitochondria-associated endoplasmic reticulum membranes (MAMs) and Ca 2+ transport in the pathogenesis of diseases. Acta Pharmacol Sin 2025; 46:271-291. [PMID: 39117969 PMCID: PMC11756407 DOI: 10.1038/s41401-024-01359-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Mitochondria and the endoplasmic reticulum (ER) are vital organelles that influence various cellular physiological and pathological processes. Recent evidence shows that about 5%-20% of the mitochondrial outer membrane is capable of forming a highly dynamic physical connection with the ER, maintained at a distance of 10-30 nm. These interconnections, known as MAMs, represent a relatively conserved structure in eukaryotic cells, acting as a critical platform for material exchange between mitochondria and the ER to maintain various aspects of cellular homeostasis. Particularly, ER-mediated Ca2+ release and recycling are intricately associated with the structure and functionality of MAMs. Thus, MAMs are integral in intracellular Ca2+ transport and the maintenance of Ca2+ homeostasis, playing an essential role in various cellular activities including metabolic regulation, signal transduction, autophagy, and apoptosis. The disruption of MAMs observed in certain pathologies such as cardiovascular and neurodegenerative diseases as well as cancers leads to a disturbance in Ca2+ homeostasis. This imbalance potentially aggravates pathological alterations and disease progression. Consequently, a thorough understanding of the link between MAM-mediated Ca2+ transport and these diseases could unveil new perspectives and therapeutic strategies. This review focuses on the changes in MAMs function during disease progression and their implications in relation to MAM-associated Ca2+ transport.
Collapse
Affiliation(s)
- Wen-Bin Zhao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
2
|
Liu J, Xu S, Gao B, Yuan M, Zhong L, Guo R. Protective effect of SERCA2a-SUMOylation by SUMO-1 on diabetes-induced atherosclerosis and aortic vascular injury. Mol Cell Biochem 2025; 480:279-293. [PMID: 38438822 DOI: 10.1007/s11010-024-04953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/26/2024] [Indexed: 03/06/2024]
Abstract
Diabetes is a major risk factor for cardiovascular disease. However, the exact mechanism by which diabetes contributes to vascular damage is not fully understood. The aim of this study was to investigate the role of SUMO-1 mediated SERCA2a SUMOylation in the development of atherosclerotic vascular injury associated with diabetes mellitus. ApoE-/- mice were treated with streptozotocin (STZ) injection combined with high-fat feeding to simulate diabetic atherosclerosis and vascular injury. Human aortic vascular smooth muscle cells (HAVSMCs) were treated with high glucose (HG, 33.3 mM) and palmitic acid (PA, 200 µM) for 24 h to mimic a model of diabetes-induced vascular injury in vitro. Aortic vascular function, phenotypic conversion, migration, proliferation, intracellular Ca2+ concentration, the levels of small ubiquitin-like modifier type 1 (SUMO1), SERCA2a and SUMOylated SERCA2a were detected. Diabetes-induced atherosclerotic mice presented obvious atherosclerotic plaques and vascular injury, companied by significantly lower levels of SUMO1 and SERCA2a in aorta. HG and PA treatment in HAVSMCs reduced the expressions of SUMO1, SERCA2a and SUMOylated SERCA2a, facilitated the HAVSMCs phenotypic transformation, proliferation and migration, attenuated the Ca2+ transport, and increased the resting intracellular Ca2+ concentration. We also confirmed that SUMO1 directly bound to SERCA2a in HAVSMCs. Overexpression of SUMO1 restored the function and phenotypic contractile ability of HAVSMCs by upregulating SERCA2a SUMOylation, thereby alleviating HG and PA-induced vascular injury. These observations suggest an essential role of SUMO1 to protect diabetes-induced atherosclerosis and aortic vascular injury by the regulation of SERCA2a-SUMOylation and calcium homeostasis.
Collapse
MESH Headings
- Animals
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- SUMO-1 Protein/metabolism
- Sumoylation
- Mice
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Humans
- Aorta/pathology
- Aorta/metabolism
- Male
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout, ApoE
Collapse
Affiliation(s)
- Jinlin Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Shifang Xu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Bin Gao
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Meng Yuan
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China.
- The Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, China.
| |
Collapse
|
3
|
Caillard P, Bennis Y, Boudot C, Chatelain D, Rybarczyk P, Boullier A, Poirot S, Titeca-Beauport D, Bodeau S, Choukroun G, Kamel S, Six I, Maizel J. Acute kidney disease in mice is associated with early cardiovascular dysfunction. Ren Fail 2024; 46:2415510. [PMID: 39422224 PMCID: PMC11492403 DOI: 10.1080/0886022x.2024.2415510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are major health concerns due to their increasing incidence and high mortality. They are interconnected syndromes; AKI without recovery evolves into acute kidney disease (AKD), which can indicate an AKI-to-CKD transition. Both AKI and CKD are associated with a risk of long-term cardiovascular complications, but whether vascular and cardiac dysfunctions can occur as early as the AKD period has not been studied extensively. In a mouse model of kidney injury (KI) with non-recovery, we performed vasoreactivity and echocardiography analyses on days 15 (D15) and 45 (D45) after KI. We determined the concentrations of two major gut-derived protein-bound uremic toxins known to induce cardiovascular toxicity-indoxyl sulfate (IS) and para-cresyl sulfate (PCS)-and the levels of inflammation and contraction markers on D7, D15, and D45. Mice with KI showed acute tubular and interstitial kidney lesions on D7 and D15 and chronic glomerulosclerosis on D45. They showed significant impairment of aorta relaxation and systolic-diastolic heart function, both on D15 and D45. Such dysfunction was associated with downregulation of the expression of two contractile proteins, αSMA and SERCA2a, with a more pronounced effect on D15 than on D45. KI was also followed by a rapid increase in IS and PCS serum concentrations and the expression induction of pro-inflammatory cytokines and endothelial adhesion molecules in serum and cardiovascular tissues. Therefore, these results highlight that AKD leads to early cardiac and vascular dysfunctions. How these dysfunctions could be managed to prevent cardiovascular events deserves further study.
Collapse
Affiliation(s)
- Pauline Caillard
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, Amiens, France
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Youssef Bennis
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
- Department of Clinical Pharmacology, Amiens Medical Center, Amiens, France
| | - Cédric Boudot
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Denis Chatelain
- Department of Anatomopathology, Amiens Medical Center, Amiens, France
| | - Pierre Rybarczyk
- Hauts-de-France Anatomopathology Institute (i-PatH), Amiens, France
| | - Agnès Boullier
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
- Department of Clinical Biochemistry, Amiens Medical Center, Amiens, France
| | - Sabrina Poirot
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Dimitri Titeca-Beauport
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, Amiens, France
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Sandra Bodeau
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
- Department of Clinical Pharmacology, Amiens Medical Center, Amiens, France
| | - Gabriel Choukroun
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, Amiens, France
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Saïd Kamel
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
- Department of Clinical Biochemistry, Amiens Medical Center, Amiens, France
| | - Isabelle Six
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
| | - Julien Maizel
- MP3CV laboratory, UR UPJV 7517, University of Picardy Jules Verne, Amiens, France
- Department of Intensive Care Medicine, Amiens Medical Center, Amiens, France
| |
Collapse
|
4
|
Heger V, Benesova B, Majekova M, Rezbarikova P, Hunyadi A, Horakova L, Viskupicova J. Polyphenolic Compounds Activate SERCA1a and Attenuate Methylglyoxal- and Palmitate-Induced Impairment in Pancreatic INS-1E Beta Cells. Cells 2024; 13:1860. [PMID: 39594609 PMCID: PMC11593225 DOI: 10.3390/cells13221860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) is an important regulatory protein responsible for maintaining calcium homeostasis within cells. Impairment of SERCA associated with activity/expression decrease has been implicated in multiple chronic conditions, including cardiovascular diseases, diabetes, cancer, neurodegenerative diseases, and skeletal muscle pathologies. Natural polyphenols have been recognized to interact with several target proteins involving SERCA. To date, only a limited number of polyphenolic compounds or their derivatives have been described either to increase SERCA activity/expression directly or to affect Ca2+ signaling pathways. In this study, we tested polyphenols for their ability to activate SERCA1a in the absence or presence of methylglyoxal or palmitate and to impact insulin release in pancreatic beta cells. The protective effects of these compounds against methylglyoxal- or palmitate-induced injury were evaluated. Results indicate that 6-gingerol, resveratrol, and ellagic acid activate SERCA1a and protect against activity decrease induced by methylglyoxal and palmitate. Molecular docking analysis revealed the binding of these polyphenols to Glu439 in the SERCA1a P-domain, suggesting a critical role in the stimulation of enzyme activity. Ellagic acid was found to directly stimulate the activity of SERCA1a, marking the first instance of such an observation.
Collapse
Affiliation(s)
- Vladimir Heger
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Barbora Benesova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
- Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia
| | - Magdalena Majekova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Petronela Rezbarikova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, H-6720 Szeged, Hungary;
| | - Lubica Horakova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Jana Viskupicova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| |
Collapse
|
5
|
Hirakis SP, Bartol TM, Autin L, Amaro RE, Sejnowski TJ. Electrophysical cardiac remodeling at the molecular level: Insights into ryanodine receptor activation and calcium-induced calcium release from a stochastic explicit-particle model. Biophys J 2024; 123:3812-3831. [PMID: 39369273 PMCID: PMC11560313 DOI: 10.1016/j.bpj.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024] Open
Abstract
We present the first-ever, fully discrete, stochastic model of triggered cardiac Ca2+ dynamics. Using anatomically accurate subcellular cardiac myocyte geometries, we simulate the molecular players involved in Ca2+ handling using high-resolution stochastic and explicit-particle methods at the level of an individual cardiac dyadic junction. Integrating data from multiple experimental sources, the model not only replicates the findings of traditional in silico studies and complements in vitro experimental data but also reveals new insights into the molecular mechanisms driving cardiac dysfunction under stress and disease conditions. We improve upon older, nondiscrete models using the same realistic geometry by incorporating molecular mechanisms for spontaneous, as well as triggered calcium-induced calcium release (CICR). Action potentials are used to activate L-type calcium channels (LTCC), triggering CICR through ryanodine receptors (RyRs) on the surface of the sarcoplasmic reticulum. These improvements allow for the specific focus on the couplon: the structure-function relationship between LTCC and RyR. We investigate the electrophysical effects of normal and diseased action potentials on CICR and interrogate the effects of dyadic junction deformation through detubulation and orphaning of RyR. Our work demonstrates the importance of the electrophysical integrity of the calcium release unit on CICR fidelity, giving insights into the molecular basis of heart disease. Finally, we provide a unique, detailed, molecular view of the CICR process using advanced rendering techniques. This easy-to-use model comes complete with tutorials and the necessary software for use and analysis to maximize usability and reproducibility. Our work focuses on quantifying, qualifying, and visualizing the behavior of the molecular species that underlie the function and dysfunction of subcellular cardiomyocyte systems.
Collapse
Affiliation(s)
- Sophia P Hirakis
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California
| | - Thomas M Bartol
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California
| | - Ludovic Autin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| | - Terrence J Sejnowski
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| |
Collapse
|
6
|
Barry C, Rouhana S, Braun JL, Geromella MS, Fajardo VA, Pyle WG. Perimenopause Decreases SERCA2a Activity in the Hearts of a Mouse Model of Ovarian Failure. Biomolecules 2024; 14:675. [PMID: 38927078 PMCID: PMC11201532 DOI: 10.3390/biom14060675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Risk of cardiovascular disease mortality rises in women after menopause. While increased cardiovascular risk is largely attributed to postmenopausal declines in estrogens, the molecular changes in the heart that contribute to risk are poorly understood. Disruptions in intracellular calcium handling develop in ovariectomized mice and have been implicated in cardiac dysfunction. Using a mouse model of menopause in which ovarian failure occurs over 120 days, we sought to determine if perimenopause impacted calcium removal mechanisms in the heart and identify the molecular mechanisms. Mice were injected with 4-vinylcyclohexene diepoxide (VCD) to induce ovarian failure over 120 days, mimicking perimenopause. Hearts were removed at 60 and 120 days after VCD injections, representing the middle and end of perimenopause. SERCA2a function was significantly diminished at the end of perimenopause. Neither SERCA2a nor phospholamban expression changed at either time point, but phospholamban phosphorylation at S16 and T17 was dynamically altered. Intrinsic SERCA inhibitors sarcolipin and myoregulin increased >4-fold at day 60, as did the native activator DWORF. At the end of perimenopause, sarcolipin and myoregulin returned to baseline levels while DWORF was significantly reduced below controls. Sodium-calcium exchanger expression was significantly increased at the end of perimenopause. These results show that the foundation for increased cardiovascular disease mortality develops in the heart during perimenopause and that regulators of calcium handling exhibit significant fluctuations over time. Understanding the temporal development of cardiovascular risk associated with menopause and the underlying mechanisms is critical to developing interventions that mitigate the rise in cardiovascular mortality that arises after menopause.
Collapse
Affiliation(s)
- Ciara Barry
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Sarah Rouhana
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Jessica L. Braun
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Mia S. Geromella
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Val A. Fajardo
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - W. Glen Pyle
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
- Women’s Health Research Institute at BC Women’s Hospital + Health Centre, Vancouver, BC V6H 2N9, Canada
| |
Collapse
|
7
|
Mitronova GY, Quentin C, Belov VN, Wegener JW, Kiszka KA, Lehnart SE. 1,4-Benzothiazepines with Cyclopropanol Groups and Their Structural Analogues Exhibit Both RyR2-Stabilizing and SERCA2a-Stimulating Activities. J Med Chem 2023; 66:15761-15775. [PMID: 37991191 PMCID: PMC10726367 DOI: 10.1021/acs.jmedchem.3c01235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
To discover new multifunctional agents for the treatment of cardiovascular diseases, we designed and synthesized a series of compounds with a cyclopropyl alcohol moiety and evaluated them in biochemical assays. Biological screening identified derivatives with dual activity: preventing Ca2+ leak through ryanodine receptor 2 (RyR2) and enhancing cardiac sarco-endoplasmic reticulum (SR) Ca2+ load by activation of Ca2+-dependent ATPase 2a (SERCA2a). The compounds that stabilize RyR2 at micro- and nanomolar concentrations are either structurally related to RyR-stabilizing drugs or Rycals or have structures similar to them. The novel compounds also demonstrate a good ability to increase ATP hydrolysis mediated by SERCA2a activity in cardiac microsomes, e.g., the half-maximal effective concentration (EC50) was as low as 383 nM for compound 12a, which is 1,4-benzothiazepine with two cyclopropanol groups. Our findings indicate that these derivatives can be considered as new lead compounds to improve cardiac function in heart failure.
Collapse
Affiliation(s)
- Gyuzel Y. Mitronova
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| | - Christine Quentin
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Vladimir N. Belov
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Jörg W. Wegener
- Department
of Cardiology & Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Strasse 42a, Göttingen 37075, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| | - Kamila A. Kiszka
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Stephan E. Lehnart
- Department
of Cardiology & Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Strasse 42a, Göttingen 37075, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| |
Collapse
|
8
|
Zawieja SD, Pea GA, Broyhill SE, Patro A, Bromert KH, Li M, Norton CE, Castorena-Gonzalez JA, Hancock EJ, Bertram CD, Davis MJ. IP3R1 underlies diastolic ANO1 activation and pressure-dependent chronotropy in lymphatic collecting vessels. J Gen Physiol 2023; 155:e202313358. [PMID: 37851027 PMCID: PMC10585095 DOI: 10.1085/jgp.202313358] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023] Open
Abstract
Pressure-dependent chronotropy of murine lymphatic collecting vessels relies on the activation of the Ca2+-activated chloride channel encoded by Anoctamin 1 (Ano1) in lymphatic muscle cells. Genetic ablation or pharmacological inhibition of ANO1 results in a significant reduction in basal contraction frequency and essentially complete loss of pressure-dependent frequency modulation by decreasing the rate of the diastolic depolarization phase of the ionic pacemaker in lymphatic muscle cells (LMCs). Oscillating Ca2+ release from sarcoendoplasmic reticulum Ca2+ channels has been hypothesized to drive ANO1 activity during diastole, but the source of Ca2+ for ANO1 activation in smooth muscle remains unclear. Here, we investigated the role of the inositol triphosphate receptor 1 (Itpr1; Ip3r1) in this process using pressure myography, Ca2+ imaging, and membrane potential recordings in LMCs of ex vivo pressurized inguinal-axillary lymphatic vessels from control or Myh11CreERT2;Ip3r1fl/fl (Ip3r1ismKO) mice. Ip3r1ismKO vessels had significant reductions in contraction frequency and tone but an increased contraction amplitude. Membrane potential recordings from LMCs of Ip3r1ismKO vessels revealed a depressed diastolic depolarization rate and an elongation of the plateau phase of the action potential (AP). Ca2+ imaging of LMCs using the genetically encoded Ca2+ sensor GCaMP6f demonstrated an elongation of the Ca2+ flash associated with an AP-driven contraction. Critically, diastolic subcellular Ca2+ transients were absent in LMCs of Ip3r1ismKO mice, demonstrating the necessity of IP3R1 activity in controlling ANO1-mediated diastolic depolarization. These findings indicate a critical role for IP3R1 in lymphatic vessel pressure-dependent chronotropy and contractile regulation.
Collapse
Affiliation(s)
- Scott D. Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Grace A. Pea
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Sarah E. Broyhill
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Advaya Patro
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Karen H. Bromert
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Min Li
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Charles E. Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | | | - Edward J. Hancock
- School of Mathematics and Statistics, University of Sydney, Sydney, Australia
| | | | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
9
|
Li X, La Salvia S, Liang Y, Adamiak M, Kohlbrenner E, Jeong D, Chepurko E, Ceholski D, Lopez-Gordo E, Yoon S, Mathiyalagan P, Agarwal N, Jha D, Lodha S, Daaboul G, Phan A, Raisinghani N, Zhang S, Zangi L, Gonzalez-Kozlova E, Dubois N, Dogra N, Hajjar RJ, Sahoo S. Extracellular Vesicle-Encapsulated Adeno-Associated Viruses for Therapeutic Gene Delivery to the Heart. Circulation 2023; 148:405-425. [PMID: 37409482 DOI: 10.1161/circulationaha.122.063759] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/16/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Adeno-associated virus (AAV) has emerged as one of the best tools for cardiac gene delivery due to its cardiotropism, long-term expression, and safety. However, a significant challenge to its successful clinical use is preexisting neutralizing antibodies (NAbs), which bind to free AAVs, prevent efficient gene transduction, and reduce or negate therapeutic effects. Here we describe extracellular vesicle-encapsulated AAVs (EV-AAVs), secreted naturally by AAV-producing cells, as a superior cardiac gene delivery vector that delivers more genes and offers higher NAb resistance. METHODS We developed a 2-step density-gradient ultracentrifugation method to isolate highly purified EV-AAVs. We compared the gene delivery and therapeutic efficacy of EV-AAVs with an equal titer of free AAVs in the presence of NAbs, both in vitro and in vivo. In addition, we investigated the mechanism of EV-AAV uptake in human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and mouse models in vivo using a combination of biochemical techniques, flow cytometry, and immunofluorescence imaging. RESULTS Using cardiotropic AAV serotypes 6 and 9 and several reporter constructs, we demonstrated that EV-AAVs deliver significantly higher quantities of genes than AAVs in the presence of NAbs, both to human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and to mouse hearts in vivo. Intramyocardial delivery of EV-AAV9-sarcoplasmic reticulum calcium ATPase 2a to infarcted hearts in preimmunized mice significantly improved ejection fraction and fractional shortening compared with AAV9-sarcoplasmic reticulum calcium ATPase 2a delivery. These data validated NAb evasion by and therapeutic efficacy of EV-AAV9 vectors. Trafficking studies using human induced pluripotent stem cell-derived cells in vitro and mouse hearts in vivo showed significantly higher expression of EV-AAV6/9-delivered genes in cardiomyocytes compared with noncardiomyocytes, even with comparable cellular uptake. Using cellular subfraction analyses and pH-sensitive dyes, we discovered that EV-AAVs were internalized into acidic endosomal compartments of cardiomyocytes for releasing and acidifying AAVs for their nuclear uptake. CONCLUSIONS Together, using 5 different in vitro and in vivo model systems, we demonstrate significantly higher potency and therapeutic efficacy of EV-AAV vectors compared with free AAVs in the presence of NAbs. These results establish the potential of EV-AAV vectors as a gene delivery tool to treat heart failure.
Collapse
Affiliation(s)
- Xisheng Li
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sabrina La Salvia
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yaxuan Liang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China (Y.L.)
| | - Marta Adamiak
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Erik Kohlbrenner
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
- Spark Therapeutics, Philadelphia, PA (E.K.)
| | - Dongtak Jeong
- Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea (D.J.)
| | - Elena Chepurko
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Delaine Ceholski
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Estrella Lopez-Gordo
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Seonghun Yoon
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Neha Agarwal
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Divya Jha
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shweta Lodha
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Anh Phan
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nikhil Raisinghani
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shihong Zhang
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lior Zangi
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Edgar Gonzalez-Kozlova
- Department of Oncological Sciences (E.G.-K.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology (N. Dubois), Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute (N. Dubois), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Navneet Dogra
- Department of Pathology and Laboratory Medicine (N. Dogra), Icahn School of Medicine at Mount Sinai, New York, NY
- Icahn Genomics Institute (N.Dogra), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- Gene and Cell Therapy Institute, Massachusetts General Brigham, Boston (R.J.H.)
| | - Susmita Sahoo
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
10
|
Gonnot F, Boulogne L, Brun C, Dia M, Gouriou Y, Bidaux G, Chouabe C, Crola Da Silva C, Ducreux S, Pillot B, Kaczmarczyk A, Leon C, Chanon S, Perret C, Sciandra F, Dargar T, Gache V, Farhat F, Sebbag L, Bochaton T, Thibault H, Ovize M, Paillard M, Gomez L. SERCA2 phosphorylation at serine 663 is a key regulator of Ca 2+ homeostasis in heart diseases. Nat Commun 2023; 14:3346. [PMID: 37291092 PMCID: PMC10250397 DOI: 10.1038/s41467-023-39027-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
Despite advances in cardioprotection, new therapeutic strategies capable of preventing ischemia-reperfusion injury of patients are still needed. Here, we discover that sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA2) phosphorylation at serine 663 is a clinical and pathophysiological event of cardiac function. Indeed, the phosphorylation level of SERCA2 at serine 663 is increased in ischemic hearts of patients and mouse. Analyses on different human cell lines indicate that preventing serine 663 phosphorylation significantly increases SERCA2 activity and protects against cell death, by counteracting cytosolic and mitochondrial Ca2+ overload. By identifying the phosphorylation level of SERCA2 at serine 663 as an essential regulator of SERCA2 activity, Ca2+ homeostasis and infarct size, these data contribute to a more comprehensive understanding of the excitation/contraction coupling of cardiomyocytes and establish the pathophysiological role and the therapeutic potential of SERCA2 modulation in acute myocardial infarction, based on the hotspot phosphorylation level of SERCA2 at serine 663 residue.
Collapse
Affiliation(s)
- Fabrice Gonnot
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Laura Boulogne
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Camille Brun
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Maya Dia
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Yves Gouriou
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Gabriel Bidaux
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Christophe Chouabe
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Claire Crola Da Silva
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Sylvie Ducreux
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Bruno Pillot
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Andrea Kaczmarczyk
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Christelle Leon
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Stephanie Chanon
- Laboratoire CarMeN, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, Functional Lipidomic Plateform, Lyon, France
| | - Coralie Perret
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Franck Sciandra
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Tanushri Dargar
- Institut NeuroMyoGène INMG-PNMG, CNRS UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Vincent Gache
- Institut NeuroMyoGène INMG-PNMG, CNRS UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Fadi Farhat
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
- Hôpital Louis Pradel, Hospices Civils de Lyon, 59 boulevard Pinel, F-69500, Bron, France
- Cardiac Surgery Department, Hospices Civils de Lyon, Hôpital Louis Pradel, 69500, Bron, France
| | - Laurent Sebbag
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
- Hôpital Louis Pradel, Hospices Civils de Lyon, 59 boulevard Pinel, F-69500, Bron, France
- Heart Failure and Transplant Department, Hospices Civils de Lyon, Hôpital Louis Pradel, 69500, Bron, France
| | - Thomas Bochaton
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
- Hôpital Louis Pradel, Hospices Civils de Lyon, 59 boulevard Pinel, F-69500, Bron, France
| | - Helene Thibault
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
- Hôpital Louis Pradel, Hospices Civils de Lyon, 59 boulevard Pinel, F-69500, Bron, France
| | - Michel Ovize
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
- Hôpital Louis Pradel, Hospices Civils de Lyon, 59 boulevard Pinel, F-69500, Bron, France
| | - Melanie Paillard
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France
| | - Ludovic Gomez
- Laboratoire CarMeN - IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500, Bron, France.
| |
Collapse
|
11
|
Yang HM, Lee JE, Kim JY, You J, Kim J, Lee HS, Yoo HM, Kong MG, Han JK, Cho HJ, Park KW, Kang HJ, Koo BK, Park YB, Kim HS. Identification of cell-biologic mechanisms of coronary artery spasm and its ex vivo diagnosis using peripheral blood-derived iPSCs. Biomater Res 2023; 27:16. [PMID: 36803875 PMCID: PMC9938986 DOI: 10.1186/s40824-023-00345-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/25/2023] [Indexed: 02/20/2023] Open
Abstract
BACKGROUND Although vasospastic angina (VSA) is known to be caused by coronary artery spasm, no study has fully elucidated the exact underlying mechanism. Moreover, in order to confirm VSA, patients should undergo invasive coronary angiography with spasm provocation test. Herein, we investigated the pathophysiology of VSA using peripheral blood-derived induced pluripotent stem cells (iPSCs) and developed an ex vivo diagnostic method for VSA. METHODS AND RESULTS With 10 mL of peripheral blood from patients with VSA, we generated iPSCs and differentiated these iPSCs into target cells. As compared with vascular smooth muscle cells (VSMCs) differentiated from iPSCs of normal subjects with negative provocation test, VSA patient-specific iPSCs-derived VSMCs showed very strong contraction in response to stimulants. Moreover, VSA patient-specific VSMCs exhibited a significant increase in stimulation-induced intracellular calcium efflux (Changes in the relative fluorescence unit [ΔF/F]; Control group vs. VSA group, 2.89 ± 0.34 vs. 10.32 ± 0.51, p < 0.01), and exclusively induced a secondary or tertiary peak of calcium efflux, suggesting that those findings could be diagnostic cut-off values for VSA. The observed hyperreactivity of VSA patient-specific VSMCs were caused by the upregulation of sarco/endoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) due to its enhanced small ubiquitin-related modifier (SUMO)ylation. This increased activity of SERCA2a was reversed by treatment with ginkgolic acid, an inhibitor of SUMOylated E1 molecules (pi/µg protein; VSA group vs. VSA + ginkgolic acid, 52.36 ± 0.71 vs. 31.93 ± 1.13, p < 0.01). CONCLUSIONS Our findings showed that abnormal calcium handling in sarco/endoplasmic reticulum could be induced by the enhanced SERCA2a activity in patients with VSA, leading to spasm. Such novel mechanisms of coronary artery spasm could be useful for drug development and diagnosis of VSA.
Collapse
Affiliation(s)
- Han-Mo Yang
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joo-Eun Lee
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Ju-Young Kim
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Jihye You
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hak Seung Lee
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hee Min Yoo
- grid.410883.60000 0001 2301 0664Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, Korea
| | - Min Gyu Kong
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,grid.412678.e0000 0004 0634 1623Department of Internal Medicine, Soon Chun Hyang University Hospital, Bucheon, Korea
| | - Jung-Kyu Han
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Kyung Woo Park
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jae Kang
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Bon-Kwon Koo
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Young-Bae Park
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080, Korea. .,National Research Laboratory for Stem Cell Niche, Seoul, Korea. .,Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea. .,Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
12
|
Ushakov A, Ivanchenko V, Gagarina A. Heart Failure And Type 2 Diabetes Mellitus: Neurohumoral, Histological And Molecular Interconnections. Curr Cardiol Rev 2023; 19:e170622206132. [PMID: 35718961 PMCID: PMC10201898 DOI: 10.2174/1573403x18666220617121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) is a global healthcare burden and a leading cause of morbidity and mortality worldwide. Type 2 diabetes mellitus (T2DM) appears to be one of the major risk factors that significantly worsen HF prognosis and increase the risk of fatal cardiovascular outcomes. Despite a great knowledge of pathophysiological mechanisms involved in HF development and progression, hospitalization rates in patients with HF and concomitant T2DM remain elevated. In this review, we discuss the complex interplay between systemic neurohumoral regulation and local cardiac mechanisms participating in myocardial remodeling and HF development in T2DM with special attention to cardiomyocyte energy metabolism, mitochondrial function and calcium metabolism, cardiomyocyte hypertrophy and death, extracellular matrix remodeling.
Collapse
Affiliation(s)
- A. Ushakov
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - V. Ivanchenko
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - A. Gagarina
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
13
|
A Large-Scale High-Throughput Screen for Modulators of SERCA Activity. Biomolecules 2022; 12:biom12121789. [PMID: 36551215 PMCID: PMC9776381 DOI: 10.3390/biom12121789] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The sarco/endoplasmic reticulum Ca-ATPase (SERCA) is a P-type ion pump that transports Ca2+ from the cytosol into the endoplasmic/sarcoplasmic reticulum (ER/SR) in most mammalian cells. It is critically important in muscle, facilitating relaxation and enabling subsequent contraction. Increasing SERCA expression or specific activity can alleviate muscle dysfunction, most notably in the heart, and we seek to develop small-molecule drug candidates that activate SERCA. Therefore, we adapted an NADH-coupled assay, measuring Ca-dependent ATPase activity of SERCA, to high-throughput screening (HTS) format, and screened a 46,000-compound library of diverse chemical scaffolds. This HTS platform yielded numerous hits that reproducibly alter SERCA Ca-ATPase activity, with few false positives. The top 19 activating hits were further tested for effects on both Ca-ATPase and Ca2+ transport, in both cardiac and skeletal SR. Nearly all hits increased Ca2+ uptake in both cardiac and skeletal SR, with some showing isoform specificity. Furthermore, dual analysis of both activities identified compounds with a range of effects on Ca2+-uptake and ATPase, which fit into distinct classifications. Further study will be needed to identify which classifications are best suited for therapeutic use. These results reinforce the need for robust secondary assays and criteria for selection of lead compounds, before undergoing HTS on a larger scale.
Collapse
|
14
|
Natural Polyphenols as SERCA Activators: Role in the Endoplasmic Reticulum Stress-Related Diseases. Molecules 2022; 27:molecules27165095. [PMID: 36014327 PMCID: PMC9415898 DOI: 10.3390/molecules27165095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) is a key protein responsible for transporting Ca2+ ions from the cytosol into the lumen of the sarco/endoplasmic reticulum (SR/ER), thus maintaining Ca2+ homeostasis within cells. Accumulating evidence suggests that impaired SERCA function is associated with disruption of intracellular Ca2+ homeostasis and induction of ER stress, leading to different chronic pathological conditions. Therefore, appropriate strategies to control Ca2+ homeostasis via modulation of either SERCA pump activity/expression or relevant signaling pathways may represent a useful approach to combat pathological states associated with ER stress. Natural dietary polyphenolic compounds, such as resveratrol, gingerol, ellagic acid, luteolin, or green tea polyphenols, with a number of health-promoting properties, have been described either to increase SERCA activity/expression directly or to affect Ca2+ signaling pathways. In this review, potential Ca2+-mediated effects of the most studied polyphenols on SERCA pumps or related Ca2+ signaling pathways are summarized, and relevant mechanisms of their action on Ca2+ regulation with respect to various ER stress-related states are depicted. All data were collected using scientific search tools (i.e., Science Direct, PubMed, Scopus, and Google Scholar).
Collapse
|
15
|
Zhang M, Xu Y, Chen Y, Yan Q, Li X, Ding L, Wei T, Zeng D. Three-Dimensional Poly-(ε-Caprolactone) Nanofibrous Scaffolds Promote the Maturation of Human Pluripotent Stem Cells-Induced Cardiomyocytes. Front Cell Dev Biol 2022; 10:875278. [PMID: 35979378 PMCID: PMC9377449 DOI: 10.3389/fcell.2022.875278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Although pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have been proved to be a new platform for heart regeneration, the lack of maturity significantly hinders the clinic application. Recent researches indicate that the function of stem cell is associated with the nanoscale geometry/topography of the extracellular matrix (ECM). However, the effects of 3D nanofibrous scaffolds in maturation of iPSC-CMs still remain unclear. Thus, we explored the effects of restructuring iPSC-CMs in 3D nano-scaffolds on cell morphology, cardiac-specific structural protein, gap junction and calcium transient kinetics. Using the electrospinning technology, poly-(ε-caprolactone) (PCL) nanofibrous scaffold were constructed and iPSC-CMs were seeded into these forms. As expected, strong sarcolemmal remodeling processes and myofilament reorientation were observed in 3D nano-scaffolds culture, as well as more expression of cardiac mature proteins, such as β-MHC and MLC2v. The mature morphology of 3D-shaped iPSC-CMs leaded to enhanced calcium transient kinetics, with increased calcium peak transient amplitude and the maximum upstroke velocity (Vmax). The results revealed that the maturation of iPSC-CMs was enhanced by the electrospun 3D PCL nanofibrous scaffolds treatment. These findings also proposed a feasible strategy to improve the myocardium bioengineering by combining stem cells with scaffolds.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, China
| | - Yan Chen
- Department of Cardiology, 971th Hospital, Chinese People’s Liberation Army Navy, Qingdao, China
| | - Qinru Yan
- Department of Neurological Rehabilitation, Xi ‘an International Medical Center Hospital, Xi’an, China
| | - Xiaoli Li
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
- *Correspondence: Di Zeng,
| |
Collapse
|
16
|
Prophylactic Evidence of MSCs-Derived Exosomes in Doxorubicin/Trastuzumab-Induced Cardiotoxicity: Beyond Mechanistic Target of NRG-1/Erb Signaling Pathway. Int J Mol Sci 2022; 23:ijms23115967. [PMID: 35682646 PMCID: PMC9181089 DOI: 10.3390/ijms23115967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/06/2022] Open
Abstract
Trastuzumab (Trz) is a humanized monoclonal antibody targeting epidermal growth factor receptor 2 (HER2; ErbB2). The combined administration of Trz and doxorubicin (DOX) has shown potent anti-cancer efficacy; however, this regimen may be accompanied by severe cardiac toxicity. Mesenchymal stem cells (MSCs)-derived exosomes are nanosized vesicles that play a crucial role in cell–cell communication and have shown efficacy in the treatment of various diseases. In this study, we aim to investigate the cardioprotective effects of MSCs-derived exosomes in a DOX/Trz- mediated cardiotoxicity model, and the possible mechanisms underlying these effects are elucidated. Forty-nine male rats were randomly assigned into four groups: Group I (control); Group II (Dox/Trz); Group III (protective group); and Group IV (curative group). Cardiac hemodynamic parameters, serum markers of cardiac injury, oxidative stress indices, and cardiac histopathology were investigated. Further, transcript profile of specific cardiac tissue injury markers, apoptotic markers, and fibrotic markers were analyzed using qRT-PCR, while the protein expressions of pAkt/Akt, pERK/ERK, pJNK/JNK, pJNK/JNK, and pSTAT3/STAT3 were evaluated by ELISA. Additionally, cardiac mirR-21 and miR-26a were assessed. A combined administration of DOX/Trz disrupted redox and Ca2+ homeostasis in cardiac tissue induced myocardial fibrosis and myofibril loss and triggered cardiac DNA damage and apoptosis. This cardiotoxicity was accompanied by decreased NRG-1 mRNA expression, HER2 protein expression, and suppressed AKT and ERK phosphorylation, while triggering JNK phosphorylation. Histological and ultra-structural examination of cardiac specimens revealed features typical of cardiac tissue injury. Moreover, a significant decline in cardiac function was observed through biochemical testing of serum cardiac markers and echocardiography. In contrast, the intraperitoneal administration of MSCs-derived exosomes alleviated cardiac injury in both protective and curative protocols; however, superior effects were observed in the protective protocol. The results of the current study indicate the ability of MSCs-derived exosomes to protect from and attenuate DOX/Trz-induced cardiotoxicity. The NRG-1/HER2, MAPK, PI3K/AKT, PJNK/JNK, and PSTAT/STAT signaling pathways play roles in mediating these effects.
Collapse
|
17
|
Zhang Y, Inaba K. Structural basis of the conformational and functional regulation of human SERCA2b, the ubiquitous endoplasmic reticulum calcium pump. Bioessays 2022; 44:e2200052. [PMID: 35560336 DOI: 10.1002/bies.202200052] [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: 03/06/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Abstract
Sarco/endoplasmic reticulum Ca2+ ATPase 2b (SERCA2b), a member of the SERCA family, is expressed ubiquitously and transports Ca2+ into the sarco/endoplasmic reticulum using the energy provided by ATP binding and hydrolysis. The crystal structure of SERCA2b in its Ca2+ - and ATP-bound (E1∙2Ca2+ -ATP) state and cryo-electron microscopy (cryo-EM) structures of the protein in its E1∙2Ca2+ -ATP and Ca2+ -unbound phosphorylated (E2P) states have provided essential insights into how the overall conformation and ATPase activity of SERCA2b is regulated by the transmembrane helix 11 and the subsequent luminal extension loop, both of which are specific to this isoform. More recently, our cryo-EM analysis has revealed that SERCA2b likely adopts open and closed conformations of the cytosolic domains in the Ca2+ -bound but ATP-free (E1∙2Ca2+ ) state, and that the closed conformation represents a state immediately prior to ATP binding. This review article summarizes the unique mechanisms underlying the conformational and functional regulation of SERCA2b.
Collapse
Affiliation(s)
- Yuxia Zhang
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, Japan.,Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, Japan.,Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Japan
| |
Collapse
|
18
|
Braun JL, Messner HN, Cleverdon REG, Baranowski RW, Hamstra SI, Geromella MS, Stuart JA, Fajardo VA. Heterozygous SOD2 deletion selectively impairs SERCA function in the soleus of female mice. Physiol Rep 2022; 10:e15285. [PMID: 35581738 PMCID: PMC9114654 DOI: 10.14814/phy2.15285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) restores intracellular Ca2+ ([Ca2+ ]i ) to resting levels after muscle contraction, ultimately eliciting relaxation. SERCA pumps are highly susceptible to tyrosine (T)-nitration, impairing their ability to take up Ca2+ resulting in reduced muscle function and increased [Ca2+ ]i and cellular damage. The mitochondrial antioxidant enzyme, superoxide dismutase 2 (SOD2), converts superoxide radicals into less reactive H2 O2 . Heterozygous deletion of SOD2 (Sod2+/- ) in mice increases mitochondrial oxidative stress; however, the consequences of reduced SOD2 expression in skeletal and cardiac muscle, specifically the effect on SERCA pumps, has yet to be investigated. We obtained soleus, extensor digitorum longus (EDL), and left ventricle (LV) muscles from 6 to 7 month-old wild-type (WT) and Sod2+/- female C57BL/6J mice. Ca2+ -dependent SERCA activity assays were performed to assess SERCA function. Western blotting was conducted to examine the protein content of SERCA, phospholamban, and sarcolipin; and immunoprecipitation experiments were done to assess SERCA2a- and SERCA1a-specific T-nitration. Heterozygous SOD2 deletion did not alter SERCA1a or SERCA2a expression in the soleus or LV but reduced SERCA2a in the EDL compared with WT, though this was not statistically significant. Soleus muscles from Sod2+/- mice showed a significant reduction in SERCA's apparent affinity for Ca2+ when compared to WT, corresponding with significantly elevated SERCA2a T-nitration in the soleus. No effect was seen in the EDL or the LV. This is the first study to investigate the effects of SOD2 deficiency on muscle SERCA function and shows that it selectively impairs SERCA function in the soleus.
Collapse
Affiliation(s)
- Jessica L. Braun
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Holt N. Messner
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Department of Biological SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Riley E. G. Cleverdon
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Ryan W. Baranowski
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Sophie I. Hamstra
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Mia S. Geromella
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Jeffrey A. Stuart
- Department of Biological SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Val A. Fajardo
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
19
|
Tabasso C, Frossard MP, Ducret C, Chehade H, Mauduit C, Benahmed M, Simeoni U, Siddeek B. Transient Post-Natal Exposure to Xenoestrogens Induces Long-Term Alterations in Cardiac Calcium Signaling. TOXICS 2022; 10:toxics10030102. [PMID: 35324727 PMCID: PMC8954167 DOI: 10.3390/toxics10030102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 02/05/2023]
Abstract
Today, non-communicable disorders are widespread worldwide. Among them, cardiovascular diseases represent the main cause of death. At the origin of these diseases, exposure to challenges during developmental windows of vulnerability (peri-conception, in utero, and early infancy periods) have been incriminated. Among the challenges that have been described, endocrine disruptors are of high concern because of their omnipresence in the environment. Worrisomely, since birth, children are exposed to a significant number of endocrine disruptors. However, the role of such early exposure on long-term cardiac health is poorly described. In this context, based on a model of rats exposed postnatally and transiently to an estrogenic compound prototype (estradiol benzoate, EB), we aimed to delineate the effects on the adult heart of such transient early exposure to endocrine disruptors and identify the underlying mechanisms involved in the potential pathogenesis. We found that this transient post-natal exposure to EB induced cardiac hypertrophy in adulthood, with increased cardiomyocyte size. The evaluation of cardiac calcium signaling, through immunoblot approaches, highlighted decreased expression of the sarcoplasmic reticulum calcium ATPase 2 (SERCA2) and decreased Nuclear Factor of Activated T Cells (NFAT3) phosphorylation as a potential underlying mechanism of cardiac hypertrophy. Furthermore, the treatment of cardiomyocytes with EB in vitro induced a decrease in SERCA2 protein levels. Overall, our study demonstrates that early transient exposure to EB induces permanent cardiac alterations. Together, our data highlight SERCA2 down-regulation as a potential mechanism involved in the cardiac pathogenesis induced by EB. These results suggest programming of adult heart dysfunctions such as arrhythmia and heart failures by early exposure to endocrine disruptors and could open new perspectives for treatment and prevention.
Collapse
Affiliation(s)
- Cassandra Tabasso
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Marie-Pauline Frossard
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Camille Ducret
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Hassib Chehade
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Claire Mauduit
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, 06204 Nice, France; (C.M.); (M.B.)
| | - Mohamed Benahmed
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, 06204 Nice, France; (C.M.); (M.B.)
| | - Umberto Simeoni
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
| | - Benazir Siddeek
- Woman-Mother-Child Department, Division of Pediatrics, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland; (C.T.); (M.-P.F.); (C.D.); (H.C.); (U.S.)
- Correspondence: ; Tel.: +41-21-3143-212
| |
Collapse
|
20
|
Sharma P, Liu Chung Ming C, Wang X, Bienvenu LA, Beck D, Figtree GA, Boyle A, Gentile C. Biofabrication of advanced in vitro3D models to study ischaemic and doxorubicin-induced myocardial damage. Biofabrication 2022; 14. [PMID: 34983029 DOI: 10.1088/1758-5090/ac47d8] [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: 12/01/2021] [Accepted: 01/04/2022] [Indexed: 11/11/2022]
Abstract
Current preclinicalin vitroandin vivomodels of cardiac injury typical of myocardial infarction (MI, or heart attack) and drug induced cardiotoxicity mimic only a few aspects of these complex scenarios. This leads to a poor translation of findings from the bench to the bedside. In this study, we biofabricated for the first time advancedin vitromodels of MI and doxorubicin (DOX) induced injury by exposing cardiac spheroids (CSs) to pathophysiological changes in oxygen (O2) levels or DOX treatment. Then, contractile function and cell death was analyzed in CSs in control versus I/R and DOX CSs. For a deeper dig into cell death analysis, 3D rendering analyses and mRNA level changes of cardiac damage-related genes were compared in control versus I/R and DOX CSs. Overall,in vitroCSs recapitulated major features typical of thein vivoMI and drug induced cardiac damages, such as adapting intracellular alterations to O2concentration changes and incubation with cardiotoxic drug, mimicking the contraction frequency and fractional shortening and changes in mRNA expression levels for genes regulating sarcomere structure, calcium transport, cell cycle, cardiac remodelling and signal transduction. Taken together, our study supports the use of I/R and DOX CSs as advancedin vitromodels to study MI and DOX-induced cardiac damage by recapitulating their complex in vivoscenario.
Collapse
Affiliation(s)
- Poonam Sharma
- The University of Newcastle Faculty of Health and Medicine, Kookaburra Cct, New Lambton Heights, New South Wales, 2305, AUSTRALIA
| | - Clara Liu Chung Ming
- University of Technology Sydney Faculty of Engineering, Building 11, Level 10, Room 115, University of Technology Sydney, Ultimo, Sydney, Ultimo, Sydney, New South Wales, 2007, AUSTRALIA
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute South Australia, 75 Commercial Road, Melbourne, Victoria, 3004, AUSTRALIA
| | - Laura A Bienvenu
- Baker Heart and Diabetes Institute South Australia, 75 Commercial Road, Melbourne, Victoria, 3004, AUSTRALIA
| | - Domink Beck
- University of Technology Sydney Faculty of Engineering, Building 11, Level 10, Room 115, University of Technology Sydney, Ultimo, Sydney, Ultimo, Sydney, New South Wales, 2007, AUSTRALIA
| | - Gemma A Figtree
- , The University of Sydney Faculty of Medicine and Health, Reserve Rd, Sydney, New South Wales, 2000, AUSTRALIA
| | - Andrew Boyle
- The University of Newcastle Faculty of Health and Medicine, Kookaburra Cct, New Lambton Heights, New South Wales, 2305, AUSTRALIA
| | - Carmine Gentile
- University of Technology Sydney Faculty of Engineering, Building 11, Level 10, Room 115, 81 Broadway St, Ultimo, Sydney, Ultimo, Sydney, New South Wales, 2007, AUSTRALIA
| |
Collapse
|
21
|
Vaiciuleviciute R, Bironaite D, Uzieliene I, Mobasheri A, Bernotiene E. Cardiovascular Drugs and Osteoarthritis: Effects of Targeting Ion Channels. Cells 2021; 10:cells10102572. [PMID: 34685552 PMCID: PMC8534048 DOI: 10.3390/cells10102572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) and cardiovascular diseases (CVD) share many similar features, including similar risk factors and molecular mechanisms. A great number of cardiovascular drugs act via different ion channels and change ion balance, thus modulating cell metabolism, osmotic responses, turnover of cartilage extracellular matrix and inflammation. These drugs are consumed by patients with CVD for many years; however, information about their effects on the joint tissues has not been fully clarified. Nevertheless, it is becoming increasingly likely that different cardiovascular drugs may have an impact on articular tissues in OA. Here, we discuss the potential effects of direct and indirect ion channel modulating drugs, including inhibitors of voltage gated calcium and sodium channels, hyperpolarization-activated cyclic nucleotide-gated channels, β-adrenoreceptor inhibitors and angiotensin-aldosterone system affecting drugs. The aim of this review was to summarize the information about activities of cardiovascular drugs on cartilage and subchondral bone and to discuss their possible consequences on the progression of OA, focusing on the modulation of ion channels in chondrocytes and other joint cells, pain control and regulation of inflammation. The implication of cardiovascular drug consumption in aetiopathogenesis of OA should be considered when prescribing ion channel modulators, particularly in long-term therapy protocols.
Collapse
Affiliation(s)
- Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA Utrecht, The Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
- Correspondence:
| |
Collapse
|
22
|
Jiang X, Yu W, Wu S, Tang L, Zhong G, Wan F, Lan J, Zhang H, Pan J, Tang Z, Zhang X, Hu L, Huang R. Arsenic (III) and/or Antimony (III) induced disruption of calcium homeostasis and endoplasmic reticulum stress resulting in apoptosis in mice heart. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112394. [PMID: 34091186 DOI: 10.1016/j.ecoenv.2021.112394] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 06/12/2023]
Abstract
Arsenic (As) and antimony (Sb) are known as an environmental contaminant with cardiotoxicity properties. The endoplasmic reticulum (ER) is the largest calcium reservoir in the cell, and its calcium homeostasis disorder plays a vital role in endoplasmic reticulum stress (ERS) and apoptosis. The objective of this study was to investigate whether As and Sb induced apoptosis via endoplasmic reticulum stress (ERS) linked to calcium homeostasis disturbance. In this study, thirty-two adult mice were gavage-fed daily with As2O3 (4 mg/kg), SbCl3 (15 mg/kg) and co-treat with SbCl3 (15 mg/kg) and As2O3 (4 mg/kg) daily for 60 days. It was observed that As or/and Sb caused histopathological lesions and ER expansion of the heart. Meanwhile, the gene expression of ER Ca2+ release channels (RyR2 and IP3R) and calmodulin-dependent protein kinase II (CaMKII) increased while the levels of mRNA and protein of ER Ca2+ uptake channel (SERCA2) downregulated significantly compared to the controls. Then, As or/and Sb induced ERS and triggered the ER apoptotic pathway by activating unfolded protein response (UPR)-associated genes ((PERK, ATF6, IRE1, XBP1, JNK, GRP78), and apoptosis-related genes (Caspase12, Caspase3, p53, CHOP). Above indicators in As + Sb group became more severe than that of As group and Sb group. Overall, our results proved that the cardiotoxicity caused by As or/and Sb might be concerning disturbing calcium homeostasis, which induced apoptosis through the ERS pathway.
Collapse
Affiliation(s)
- Xuanxuan Jiang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Wenlan Yu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Shaofeng Wu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Lixuan Tang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Gaolong Zhong
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Fang Wan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Juan Lan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Hui Zhang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Jiaqiang Pan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaoyong Zhang
- Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| | - Lianmei Hu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
23
|
Cross-Talk between Mechanosensitive Ion Channels and Calcium Regulatory Proteins in Cardiovascular Health and Disease. Int J Mol Sci 2021; 22:ijms22168782. [PMID: 34445487 PMCID: PMC8395829 DOI: 10.3390/ijms22168782] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/12/2022] Open
Abstract
Mechanosensitive ion channels are widely expressed in the cardiovascular system. They translate mechanical forces including shear stress and stretch into biological signals. The most prominent biological signal through which the cardiovascular physiological activity is initiated or maintained are intracellular calcium ions (Ca2+). Growing evidence show that the Ca2+ entry mediated by mechanosensitive ion channels is also precisely regulated by a variety of key proteins which are distributed in the cell membrane or endoplasmic reticulum. Recent studies have revealed that mechanosensitive ion channels can even physically interact with Ca2+ regulatory proteins and these interactions have wide implications for physiology and pathophysiology. Therefore, this paper reviews the cross-talk between mechanosensitive ion channels and some key Ca2+ regulatory proteins in the maintenance of calcium homeostasis and its relevance to cardiovascular health and disease.
Collapse
|
24
|
Chen CC, Chen BR, Wang Y, Curman P, Beilinson HA, Brecht RM, Liu CC, Farrell RJ, de Juan-Sanz J, Charbonnier LM, Kajimura S, Ryan TA, Schatz DG, Chatila TA, Wikstrom JD, Tyler JK, Sleckman BP. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity is required for V(D)J recombination. J Exp Med 2021; 218:212182. [PMID: 34033676 PMCID: PMC8155808 DOI: 10.1084/jem.20201708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 04/05/2021] [Accepted: 05/07/2021] [Indexed: 11/26/2022] Open
Abstract
A whole-genome CRISPR/Cas9 screen identified ATP2A2, the gene encoding the Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) 2 protein, as being important for V(D)J recombination. SERCAs are ER transmembrane proteins that pump Ca2+ from the cytosol into the ER lumen to maintain the ER Ca2+ reservoir and regulate cytosolic Ca2+-dependent processes. In preB cells, loss of SERCA2 leads to reduced V(D)J recombination kinetics due to diminished RAG-mediated DNA cleavage. SERCA2 deficiency in B cells leads to increased expression of SERCA3, and combined loss of SERCA2 and SERCA3 results in decreased ER Ca2+ levels, increased cytosolic Ca2+ levels, reduction in RAG1 and RAG2 gene expression, and a profound block in V(D)J recombination. Mice with B cells deficient in SERCA2 and humans with Darier disease, caused by heterozygous ATP2A2 mutations, have reduced numbers of mature B cells. We conclude that SERCA proteins modulate intracellular Ca2+ levels to regulate RAG1 and RAG2 gene expression and V(D)J recombination and that defects in SERCA functions cause lymphopenia.
Collapse
Affiliation(s)
- Chun-Chin Chen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Bo-Ruei Chen
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Yinan Wang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Philip Curman
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Dermato-Venereology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Ryan M Brecht
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Catherine C Liu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Ryan J Farrell
- Department of Biochemistry, Weill Cornell Medicine, New York, NY.,David Rockefeller Graduate Program, The Rockefeller University, New York, NY
| | | | | | - Shingo Kajimura
- Beth Israel Deaconess Medical Center, Division of Endocrinology, Diabetes and Metabolism, Harvard Medical School, Boston, MA
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, NY
| | - David G Schatz
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Talal A Chatila
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Boston, MA
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Dermato-Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica K Tyler
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Barry P Sleckman
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| |
Collapse
|
25
|
Pang Y, Thomas P. Involvement of sarco/endoplasmic reticulum Ca 2+-ATPase (SERCA) in mPRα (PAQR7)-mediated progesterone induction of vascular smooth muscle relaxation. Am J Physiol Endocrinol Metab 2021; 320:E453-E466. [PMID: 33427050 DOI: 10.1152/ajpendo.00359.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Progesterone acts directly on vascular smooth muscle cells (VSMCs) through activation of membrane progesterone receptor α (mPRα)-dependent signaling to rapidly decrease cytosolic Ca2+ concentrations and induce muscle relaxation. However, it is not known whether this progesterone action involves uptake of Ca2+ by the sarco/endoplasmic reticulum (SR) and increased sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. The present results show that treatment of cultured human VSMCs with progesterone and the selective mPR agonist Org OD-02-0 (OD 02-0) but not with the nuclear PR agonist R5020 increased SERCA protein expression, which was blocked by knockdown of mPRα with siRNA. Moreover, treatments with progesterone and OD 02-0, but not with R5020, increased phospholamban (PLB) phosphorylation, which would result in disinhibition of SERCA function. Progesterone and OD 02-0 significantly increased Ca2+ levels in the SR and caused VSMC relaxation. These effects were blocked by pretreatment with cyclopiazonic acid (CPA), a SERCA inhibitor, and by knockdown of SERCA2 with siRNA, suggesting that SERCA2 plays a critical role in progesterone induction of VSMC relaxation. Treatment with inhibitors of inhibitory G proteins (Gi, NF023), MAP kinase (AZD 6244), Akt/Pi3k (wortmannin), and a Rho activator (calpeptin) blocked the progesterone- and OD 02-0-induced increase in Ca2+ levels in the SR and SERCA expressions. These results suggest that the rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.NEW & NOTEWORTHY The rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.
Collapse
Affiliation(s)
- Yefei Pang
- Marine Science Institute, University of Texas at Austin, Port Aransas, Texas
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, Texas
| |
Collapse
|
26
|
Pagliaro L, Marchesini M, Roti G. Targeting oncogenic Notch signaling with SERCA inhibitors. J Hematol Oncol 2021; 14:8. [PMID: 33407740 PMCID: PMC7789735 DOI: 10.1186/s13045-020-01015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.
Collapse
Affiliation(s)
- Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Matteo Marchesini
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy.
| |
Collapse
|
27
|
Chun P. Therapeutic effects of histone deacetylase inhibitors on heart disease. Arch Pharm Res 2020; 43:1276-1296. [PMID: 33245518 DOI: 10.1007/s12272-020-01297-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 01/04/2023]
Abstract
A wide range of histone deacetylase (HDAC) inhibitors have been studied for their therapeutic potential because the excessive activity and expression of HDACs have been implicated in the pathogenesis of cardiac diseases. An increasing number of preclinical studies have demonstrated the cardioprotective effects of numerous HDAC inhibitors, suggesting a wide variety of mechanisms by which the inhibitors protect against cardiac stress, such as the suppression of cardiac fibrosis and fetal gene expression, enhancement of angiogenesis and mitochondrial biogenesis, prevention of electrical remodeling, and regulation of apoptosis, autophagy, and cell cycle arrest. For the development of isoform-selective HDAC inhibitors with high efficacy and low toxicity, it is important to identify and understand the mechanisms responsible for the effects of the inhibitors. This review highlights the preclinical effects of HDAC inhibitors that act against Zn2+-dependent HDACs and the underlying mechanisms of their protective effects against cardiac hypertrophy, hypertension, myocardial infarction, heart failure, and atrial fibrillation.
Collapse
Affiliation(s)
- Pusoon Chun
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Inje-ro, Gimhae, Gyeongnam, 50834, Republic of Korea.
| |
Collapse
|
28
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Wang R, Wang M, He S, Sun G, Sun X. Targeting Calcium Homeostasis in Myocardial Ischemia/Reperfusion Injury: An Overview of Regulatory Mechanisms and Therapeutic Reagents. Front Pharmacol 2020; 11:872. [PMID: 32581817 PMCID: PMC7296066 DOI: 10.3389/fphar.2020.00872] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Calcium homeostasis plays an essential role in maintaining excitation–contraction coupling (ECC) in cardiomyocytes, including calcium release, recapture, and storage. Disruption of calcium homeostasis may affect heart function, leading to the development of various heart diseases. Myocardial ischemia/reperfusion (MI/R) injury may occur after revascularization, which is a treatment used in coronary heart disease. MI/R injury is a complex pathological process, and the main cause of increased mortality and disability after treatment of coronary heart disease. However, current methods and drugs for treating MI/R injury are very scarce, not ideal, and have limitations. Studies have shown that MI/R injury can cause calcium overload that can further aggravate MI/R injury. Therefore, we reviewed the effects of critical calcium pathway regulators on MI/R injury and drew an intuitive diagram of the calcium homeostasis pathway. We also summarized and analyzed calcium pathway-related or MI/R drugs under research or marketing by searching Therapeutic Target and PubMed Databases. The data analysis showed that six drugs and corresponding targets are used to treat MI/R injury and involved in calcium signaling pathways. We emphasize the relevance of further detailed investigation of MI/R injury and calcium homeostasis and the therapeutic role of calcium homeostasis in MI/R injury, which bridges basic research and clinical applications of MI/R injury.
Collapse
Affiliation(s)
- Ruiying Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaibing He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
30
|
SMN-deficiency disrupts SERCA2 expression and intracellular Ca 2+ signaling in cardiomyocytes from SMA mice and patient-derived iPSCs. Skelet Muscle 2020; 10:16. [PMID: 32384912 PMCID: PMC7206821 DOI: 10.1186/s13395-020-00232-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 11/17/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by loss of alpha motor neurons and skeletal muscle atrophy. The disease is caused by mutations of the SMN1 gene that result in reduced functional expression of survival motor neuron (SMN) protein. SMN is ubiquitously expressed, and there have been reports of cardiovascular dysfunction in the most severe SMA patients and animal models of the disease. In this study, we directly assessed the function of cardiomyocytes isolated from a severe SMA model mouse and cardiomyocytes generated from patient-derived IPSCs. Consistent with impaired cardiovascular function at the very early disease stages in mice, heart failure markers such as brain natriuretic peptide were significantly elevated. Functionally, cardiomyocyte relaxation kinetics were markedly slowed and the T50 for Ca2+ sequestration increased to 146 ± 4 ms in SMN-deficient cardiomyocytes from 126 ± 4 ms in wild type cells. Reducing SMN levels in cardiomyocytes from control patient IPSCs slowed calcium reuptake similar to SMA patent-derived cardiac cells. Importantly, restoring SMN increased calcium reuptake rate. Taken together, these results indicate that SMN deficiency impairs cardiomyocyte function at least partially through intracellular Ca2+ cycling dysregulation.
Collapse
|
31
|
Fransen P, Chen J, Vangheluwe P, Guns PJ. Contractile Behavior of Mouse Aorta Depends on SERCA2 Isoform Distribution: Effects of Replacing SERCA2a by SERCA2b. Front Physiol 2020; 11:282. [PMID: 32296344 PMCID: PMC7136392 DOI: 10.3389/fphys.2020.00282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/12/2020] [Indexed: 01/16/2023] Open
Abstract
The Sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) actively pumps Ca2+ into the sarco/endoplasmic reticulum, thereby regulating intracellular Ca2+ concentrations and associated physiological processes. Different SERCA isoforms have been described (SERCA1, 2, and 3) with SERCA2 playing a pivotal role in Ca2+ homeostasis in cardiovascular tissues. In the heart, SERCA2a is the dominant isoform and has been proposed as therapeutic target in patients with heart failure. In the vasculature, both SERCA2a and SERCA2b are expressed with SERCA2b being the predominant isoform. The physiological role of SERCA2a in the vasculature, however, remains incompletely understood. In the present study, we used gene-modified mice in which the alternative splicing of the SERCA2-encoding gene (Atp2a2), underlying the expression of SERCA2a, is prevented and SERCA2a is replaced by SERCA2b. The resulting SERCA2b/b mice provide a unique opportunity to investigate the specific contribution of SERCA2a versus SERCA2b to vascular physiology. Aortic segments of SERCA2b/b (SERCA2a-deficient) and SERCA2a/b (control) mice were mounted in organ baths to evaluate vascular reactivity. SERCA2b/b aortic rings displayed higher contractions induced by phenylephrine (1 μM). Surprisingly, the initial inositol-3-phosphate mediated phasic contraction showed a faster decay of force in SERCA2b/b mice, while the subsequent tonic contraction was larger in SERCA2b/b segments. Moreover, in the presence of the calcium channel blocker diltiazem (35 μM) SERCA2b/b aortic rings showed higher contractions compared to SERCA2a/b, suggesting that SERCA2a (deficiency) modulates the activity of non-selective cation channels. Additionally, in endothelial cell (EC)-denuded aortic segments, the SERCA-inhibitor cyclopiazonic acid (CPA) caused markedly larger contractions in SERCA2b/b mice, while the increases of cytosolic Ca2+ were similar in both strains. Hence, aortas of SERCA2b/b mice appear to have a stronger coupling of intracellular Ca2+ to contraction, which may be in agreement with the reported difference in intracellular localization of SERCA2a versus SERCA2b. Finally, EC-mediated relaxation by acetylcholine and ATP was assessed. Concentration-response-curves for ATP showed a higher sensitivity of aortic segments of SERCA2b/b mice, while no difference in potency between strains were observed for acetylcholine. In summary, despite the relative low expression of SERCA2a in the murine aorta, our results point toward a distinct role in vascular physiology.
Collapse
Affiliation(s)
- Paul Fransen
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Jialin Chen
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
32
|
Silbernagel N, Körner A, Balitzki J, Jaggy M, Bertels S, Richter B, Hippler M, Hellwig A, Hecker M, Bastmeyer M, Ullrich ND. Shaping the heart: Structural and functional maturation of iPSC-cardiomyocytes in 3D-micro-scaffolds. Biomaterials 2020; 227:119551. [DOI: 10.1016/j.biomaterials.2019.119551] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/06/2019] [Accepted: 10/14/2019] [Indexed: 02/05/2023]
|
33
|
Danese A, Marchi S, Vitto VAM, Modesti L, Leo S, Wieckowski MR, Giorgi C, Pinton P. Cancer-Related Increases and Decreases in Calcium Signaling at the Endoplasmic Reticulum-Mitochondria Interface (MAMs). Rev Physiol Biochem Pharmacol 2020; 185:153-193. [PMID: 32789789 DOI: 10.1007/112_2020_43] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER)-mitochondria regions are specialized subdomains called also mitochondria-associated membranes (MAMs). MAMs allow regulation of lipid synthesis and represent hubs for ion and metabolite signaling. As these two organelles can module both the amplitude and the spatiotemporal patterns of calcium (Ca2+) signals, this particular interaction controls several Ca2+-dependent pathways well known for their contribution to tumorigenesis, such as metabolism, survival, sensitivity to cell death, and metastasis. Mitochondria-mediated apoptosis arises from mitochondrial Ca2+ overload, permeabilization of the mitochondrial outer membrane, and the release of mitochondrial apoptotic factors into the cytosol. Decreases in Ca2+ signaling at the ER-mitochondria interface are being studied in depth as failure of apoptotic-dependent cell death is one of the predominant characteristics of cancer cells. However, some recent papers that linked MAMs Ca2+ crosstalk-related upregulation to tumor onset and progression have aroused the interest of the scientific community.In this review, we will describe how different MAMs-localized proteins modulate the effectiveness of Ca2+-dependent apoptotic stimuli by causing both increases and decreases in the ER-mitochondria interplay and, specifically, by modulating Ca2+ signaling.
Collapse
Affiliation(s)
- Alberto Danese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Modesti
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Sara Leo
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
34
|
Affiliation(s)
- Stuart G Campbell
- From the Department of Biomedical Engineering (S.G.C.), Yale University, New Haven, CT.,Department of Cellular and Molecular Physiology (S.G.C.), Yale School of Medicine, New Haven, CT
| | - Yibing Qyang
- From the Department of Biomedical Engineering (S.G.C.), Yale University, New Haven, CT.,Yale Stem Cell Center (Y.Q.), Yale University, New Haven, CT.,Vascular Biology and Therapeutics Program (Y.Q.), Yale University, New Haven, CT.,Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine (Y.Q.), Yale School of Medicine, New Haven, CT
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT (J.T.H.).,Department of Cardiology, UConn Health, Farmington, CT (J.T.H.)
| |
Collapse
|
35
|
Metzinger-Le Meuth V, Metzinger L. miR-223 and other miRNA's evaluation in chronic kidney disease: Innovative biomarkers and therapeutic tools. Noncoding RNA Res 2019; 4:30-35. [PMID: 30891535 PMCID: PMC6404357 DOI: 10.1016/j.ncrna.2019.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/04/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRNAs) represent a recent breakthrough regarding gene expression regulation. They are instrumental players known to regulate post-transcriptional expression. miRNAs are short single stranded RNAs that base-pair with target mRNAs in specific regions mainly within their 3' untranslated region. We know now that miRNAs are involved in kidney physiopathology. We outline in this review the recent discoveries made on the roles of miRNAs in cellular and animal models of kidney disease but also in patients suffering from chronic kidney disease, acute kidney injury and so forth. miRNAs are potential innovative biomarkers in nephrology, but before being used in daily clinical routine, their expression in large cohorts will have to be assessed, and an effort will have to be made to standardize measurement methods and to select the most suitable tissues and biofluids. In addition to a putative role as biomarkers, up- or down-regulating miRNAs is a novel therapeutic approach to cure kidney disorders. We discuss in this review recent methods that could be used to deliver miRNAs in a specific and suitable way in kidney and other organs damaged by kidney failure such as the cardiovascular system.
Collapse
Affiliation(s)
- Valérie Metzinger-Le Meuth
- INSERM U1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, Université Paris 13-Sorbonne Paris Cité, 93017 Bobigny Cedex, France
| | - Laurent Metzinger
- HEMATIM EA4666, C.U.R.S, Université de Picardie Jules Verne, 80025 Amiens Cedex 1, France
| |
Collapse
|
36
|
Wang R, Zhang J, Wang S, Wang M, Ye T, Du Y, Xie X, Ye J, Sun G, Sun X. The Cardiotoxicity Induced by Arsenic Trioxide is Alleviated by Salvianolic Acid A via Maintaining Calcium Homeostasis and Inhibiting Endoplasmic Reticulum Stress. Molecules 2019; 24:molecules24030543. [PMID: 30717322 PMCID: PMC6384753 DOI: 10.3390/molecules24030543] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/02/2022] Open
Abstract
Arsenic trioxide (ATO) has been verified as a breakthrough with respect to the management of acute promyelocytic leukemia (APL) in recent decades but associated with some serious adverse phenomena, particularly cardiac functional abnormalities. Salvianolic acid A (Sal A) is a major effective component in treating ATO-induced cardiotoxicity. Therefore, the objective of our study was to assess whether Sal A had protective effects by the regulation of calcium homeostasis and endoplasmic reticulum (ER) stress. For the in vivo study, BALB/c mice were treated with ATO and/or Sal A via daily tail vein injections for two weeks. For the in vitro study, we detected the effects of ATO and/or Sal A in real time using adult rat ventricular myocytes (ARVMs) and an IonOptix MyoCam system. Our results showed that Sal A pretreatment alleviated cardiac dysfunction and Ca2+ overload induced by ATO in vivo and vitro. Moreover, Sal A increased sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA) activity and expression, alleviated [Ca2+]ER depletion, and decreased ER stress-related protein expression. Sal A protects the heart from ATO-induced injury and its administration correlates with the modulation of SERCA, the recovery of Ca2+ homeostasis, and the down-regulation of ER stress-mediated apoptosis.
Collapse
Affiliation(s)
- Ruiying Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Jingyi Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Tianyuan Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Yuyang Du
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Xueheng Xie
- Harbin University of Commerce, Harbin 150028, China.
| | - Jingxue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| |
Collapse
|
37
|
Cha HH, Hwang JR, Sung JH, Choi SJ, Oh SY, Roh CR. Changes in calcium channel proteins according to magnesium sulfate administration in placentas from pregnancies with pre-eclampsia or fetal growth restriction. J Investig Med 2018; 67:319-326. [PMID: 30415221 PMCID: PMC6581081 DOI: 10.1136/jim-2018-000844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2018] [Indexed: 11/06/2022]
Abstract
We aimed to evaluate the changes in plasma membrane Ca2+-ATPase (PMCA) and sarcoendoplasmic reticulum CA2+-ATPase (SERCA-2) according to the antepartal magnesium sulfate (MgSO4) administration in the placentas from pregnancies with pre-eclampsia (PE) or fetal growth restriction (FGR). Pregnant women were classified as follows: (group 1) pregnancies without PE or FGR (n=16), (group 2) pregnancies with PE or FGR but without MgSO4 administration (n=14), and (group 3) pregnancies with PE or FGR and with MgSO4 administration (n=28). We observed the localization of PMCA and SERCA-2 in placentas and compared its expression among 3 groups. And we observed its expression in BeWo cells following treatment with MgSO4 and CoCl2. PMCA staining was more observed in the basal membrane, whereas SERCA-2 staining was observed predominantly under the microvillous membrane. SERCA-2 expression was significantly increased in group 3 compared with that in group 1. Considering the gestational age at delivery, PMCA expression was increased in group 2 and group 3 compared with that in group 1 after 36 weeks of gestation. SERCA-2 was increased in group 3, but not in group 2 compared with that in group 1 after 36 weeks of gestation. In BeWo cells, MgSO4 treatment increased PMCA and SERCA-2 expression. PMCA expression was influenced by gestational age at delivery, and SERCA-2 expression was increased in the presence of PE and antepartal MgSO4 administration. This indicates that antepartal MgSO4 administration has a greater influence on SERCA-2 than PMCA.
Collapse
Affiliation(s)
- Hyun-Hwa Cha
- Department of Obstetrics and Gynecology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jae-Ryoung Hwang
- Sungkynkwan University School of Medicine, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Ji Hee Sung
- Department of Obstetrics and Gynecology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheong-Rae Roh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Skogestad J, Aronsen JM. Hypokalemia-Induced Arrhythmias and Heart Failure: New Insights and Implications for Therapy. Front Physiol 2018; 9:1500. [PMID: 30464746 PMCID: PMC6234658 DOI: 10.3389/fphys.2018.01500] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
Routine use of diuretics and neurohumoral activation make hypokalemia (serum K+ < 3. 5 mM) a prevalent electrolyte disorder among heart failure patients, contributing to the increased risk of ventricular arrhythmias and sudden cardiac death in heart failure. Recent experimental studies have suggested that hypokalemia-induced arrhythmias are initiated by the reduced activity of the Na+/K+-ATPase (NKA), subsequently leading to Ca2+ overload, Ca2+/Calmodulin-dependent kinase II (CaMKII) activation, and development of afterdepolarizations. In this article, we review the current mechanistic evidence of hypokalemia-induced triggered arrhythmias and discuss how molecular changes in heart failure might lower the threshold for these arrhythmias. Finally, we discuss how recent insights into hypokalemia-induced arrhythmias could have potential implications for future antiarrhythmic treatment strategies.
Collapse
Affiliation(s)
- Jonas Skogestad
- Division of Cardiovascular and Pulmonary Diseases, Institute of Experimental Medical Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.,Bjørknes College, Oslo, Norway
| |
Collapse
|
39
|
Liu G, Li SQ, Hu PP, Tong XY. Altered sarco(endo)plasmic reticulum calcium adenosine triphosphatase 2a content: Targets for heart failure therapy. Diab Vasc Dis Res 2018; 15:322-335. [PMID: 29762054 DOI: 10.1177/1479164118774313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Sarco(endo)plasmic reticulum calcium adenosine triphosphatase is responsible for transporting cytosolic calcium into the sarcoplasmic reticulum and endoplasmic reticulum to maintain calcium homeostasis. Sarco(endo)plasmic reticulum calcium adenosine triphosphatase is the dominant isoform expressed in cardiac tissue, which is regulated by endogenous protein inhibitors, post-translational modifications, hormones as well as microRNAs. Dysfunction of sarco(endo)plasmic reticulum calcium adenosine triphosphatase is associated with heart failure, which makes sarco(endo)plasmic reticulum calcium adenosine triphosphatase a promising target for heart failure therapy. This review summarizes current approaches to ameliorate sarco(endo)plasmic reticulum calcium adenosine triphosphatase function and focuses on phospholamban, an endogenous inhibitor of sarco(endo)plasmic reticulum calcium adenosine triphosphatase, pharmacological tools and gene therapies.
Collapse
Affiliation(s)
- Gang Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Si Qi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Ping Ping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiao Yong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
40
|
Nakka K, Ghigna C, Gabellini D, Dilworth FJ. Diversification of the muscle proteome through alternative splicing. Skelet Muscle 2018; 8:8. [PMID: 29510724 PMCID: PMC5840707 DOI: 10.1186/s13395-018-0152-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/15/2018] [Indexed: 12/16/2022] Open
Abstract
Background Skeletal muscles express a highly specialized proteome that allows the metabolism of energy sources to mediate myofiber contraction. This muscle-specific proteome is partially derived through the muscle-specific transcription of a subset of genes. Surprisingly, RNA sequencing technologies have also revealed a significant role for muscle-specific alternative splicing in generating protein isoforms that give specialized function to the muscle proteome. Main body In this review, we discuss the current knowledge with respect to the mechanisms that allow pre-mRNA transcripts to undergo muscle-specific alternative splicing while identifying some of the key trans-acting splicing factors essential to the process. The importance of specific splicing events to specialized muscle function is presented along with examples in which dysregulated splicing contributes to myopathies. Though there is now an appreciation that alternative splicing is a major contributor to proteome diversification, the emergence of improved “targeted” proteomic methodologies for detection of specific protein isoforms will soon allow us to better appreciate the extent to which alternative splicing modifies the activity of proteins (and their ability to interact with other proteins) in the skeletal muscle. In addition, we highlight a continued need to better explore the signaling pathways that contribute to the temporal control of trans-acting splicing factor activity to ensure specific protein isoforms are expressed in the proper cellular context. Conclusions An understanding of the signal-dependent and signal-independent events driving muscle-specific alternative splicing has the potential to provide us with novel therapeutic strategies to treat different myopathies. Electronic supplementary material The online version of this article (10.1186/s13395-018-0152-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kiran Nakka
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Claudia Ghigna
- Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), Pavia, Italy
| | - Davide Gabellini
- Unit of Gene Expression and Muscular Dystrophy, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, DIBIT2, 5A3-44, via Olgettina 58, 20132, Milan, Italy.
| | - F Jeffrey Dilworth
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada. .,Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Rd, Mailbox 511, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
41
|
Krishnan B, Massilamany C, Basavalingappa RH, Gangaplara A, Rajasekaran RA, Afzal MZ, Khalilzad-Sharghi V, Zhou Y, Riethoven JJ, Nandi SS, Mishra PK, Sobel RA, Strande JL, Steffen D, Reddy J. Epitope Mapping of SERCA2a Identifies an Antigenic Determinant That Induces Mainly Atrial Myocarditis in A/J Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:523-537. [PMID: 29229678 PMCID: PMC5760440 DOI: 10.4049/jimmunol.1701090] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/05/2017] [Indexed: 12/20/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA)2a, a critical regulator of calcium homeostasis, is known to be decreased in heart failure. Patients with myocarditis or dilated cardiomyopathy develop autoantibodies to SERCA2a suggesting that they may have pathogenetic significance. In this report, we describe epitope mapping analysis of SERCA2a in A/J mice that leads us to make five observations: 1) SERCA2a contains multiple T cell epitopes that induce varying degrees of myocarditis. One epitope, SERCA2a 971-990, induces widespread atrial inflammation without affecting noncardiac tissues; the cardiac abnormalities could be noninvasively captured by echocardiography, electrocardiography, and magnetic resonance microscopy imaging. 2) SERCA2a 971-990-induced disease was associated with the induction of CD4 T cell responses and the epitope preferentially binds MHC class II/IAk rather than IEk By creating IAk/and IEk/SERCA2a 971-990 dextramers, the T cell responses were determined by flow cytometry to be Ag specific. 3) SERCA2a 971-990-sensitized T cells produce both Th1 and Th17 cytokines. 4) Animals immunized with SERCA2a 971-990 showed Ag-specific Abs with enhanced production of IgG2a and IgG2b isotypes, suggesting that SERCA2a 971-990 can potentially act as a common epitope for both T cells and B cells. 5) Finally, SERCA2a 971-990-sensitized T cells were able to transfer disease to naive recipients. Together, these data indicate that SERCA2a is a critical autoantigen in the mediation of atrial inflammation in mice and that our model may be helpful to study the inflammatory events that underlie the development of conditions such as atrial fibrillation in humans.
Collapse
Affiliation(s)
- Bharathi Krishnan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Rakesh H Basavalingappa
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Arunakumar Gangaplara
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rajkumar A Rajasekaran
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | | | - Vahid Khalilzad-Sharghi
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588
| | | | - Shyam S Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Raymond A Sobel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304
| | | | - David Steffen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583;
| |
Collapse
|
42
|
Abstract
The calcium pump (a.k.a. Ca2+-ATPase or SERCA) is a membrane transport protein ubiquitously found in the endoplasmic reticulum (ER) of all eukaryotic cells. As a calcium transporter, SERCA maintains the low cytosolic calcium level that enables a vast array of signaling pathways and physiological processes (e.g. synaptic transmission, muscle contraction, fertilization). In muscle cells, SERCA promotes relaxation by pumping calcium ions from the cytosol into the lumen of the sarcoplasmic reticulum (SR), the main storage compartment for intracellular calcium. X-ray crystallographic studies have provided an extensive understanding of the intermediate states that SERCA populates as it progresses through the calcium transport cycle. Historically, SERCA is also known to be regulated by small transmembrane peptides, phospholamban (PLN) and sarcolipin (SLN). PLN is expressed in cardiac muscle, whereas SLN predominates in skeletal and atrial muscle. These two regulatory subunits play critical roles in cardiac contractility. While our understanding of these regulatory mechanisms are still developing, SERCA and PLN are one of the best understood examples of peptide-transporter regulatory interactions. Nonetheless, SERCA appeared to have only two regulatory subunits, while the related sodium pump (a.k.a. Na+, K+-ATPase) has at least nine small transmembrane peptides that provide tissue specific regulation. The last few years have seen a renaissance in our understanding of SERCA regulatory subunits. First, structures of the SERCA-SLN and SERCA-PLN complexes revealed molecular details of their interactions. Second, an array of micropeptides concealed within long non-coding RNAs have been identified as new SERCA regulators. This chapter will describe our current understanding of SERCA structure, function, and regulation.
Collapse
|
43
|
Ebenebe OV, Heather A, Erickson JR. CaMKII in Vascular Signalling: "Friend or Foe"? Heart Lung Circ 2017; 27:560-567. [PMID: 29409723 DOI: 10.1016/j.hlc.2017.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/21/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023]
Abstract
Signalling mechanisms within and between cells of the vasculature enable function and maintain homeostasis. However, a number of these mechanisms also contribute to the pathophysiology of vascular disease states. The multifunctional signalling molecule calcium/calmodulin-dependent kinase II (CaMKII) has been shown to have critical functional effects in many tissue types. For example, CaMKII is known to have a dual role in cardiac physiology and pathology. The function of CaMKII within the vasculature is incompletely understood, but emerging evidence points to potential physiological and pathological roles. This review discusses the evidence for CaMKII signalling within the vasculature, with the aim to better understand both positive and potentially deleterious effects of CaMKII activation in vascular tissue.
Collapse
Affiliation(s)
- Obialunanma V Ebenebe
- Department of Physiology, School of Medical Sciences and HeartOtago, University of Otago, Dunedin, Otago, New Zealand
| | - Alison Heather
- Department of Physiology, School of Medical Sciences and HeartOtago, University of Otago, Dunedin, Otago, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology, School of Medical Sciences and HeartOtago, University of Otago, Dunedin, Otago, New Zealand.
| |
Collapse
|
44
|
Renaud-Gabardos E, Tatin F, Hantelys F, Lebas B, Calise D, Kunduzova O, Masri B, Pujol F, Sicard P, Valet P, Roncalli J, Chaufour X, Garmy-Susini B, Parini A, Prats AC. Therapeutic Benefit and Gene Network Regulation by Combined Gene Transfer of Apelin, FGF2, and SERCA2a into Ischemic Heart. Mol Ther 2017; 26:902-916. [PMID: 29249393 DOI: 10.1016/j.ymthe.2017.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/06/2017] [Accepted: 11/10/2017] [Indexed: 01/16/2023] Open
Abstract
Despite considerable advances in cardiovascular disease treatment, heart failure remains a public health challenge. In this context, gene therapy appears as an attractive approach, but clinical trials using single therapeutic molecules result in moderate benefit. With the objective of improving ischemic heart failure therapy, we designed a combined treatment, aimed to simultaneously stimulate angiogenesis, prevent cardiac remodeling, and restore contractile function. We have previously validated IRES-based vectors as powerful tools to co-express genes of interest. Mono- and multicistronic lentivectors expressing fibroblast growth factor 2 (angiogenesis), apelin (cardioprotection), and/or SERCA2a (contractile function) were produced and administrated by intramyocardial injection into a mouse model of myocardial infarction. Data reveal that combined treatment simultaneously improves vessel number, heart function parameters, and fibrosis prevention, due to FGF2, SERCA2a, and apelin, respectively. Furthermore, addition of SERCA2a in the combination decreases cardiomyocyte hypertrophy. Large-scale transcriptome analysis reveals that the triple treatment is the most efficient in restoring angiogenic balance as well as expression of genes involved in cardiac function and remodeling. Our study validates the concept of combined treatment of ischemic heart disease with apelin, FGF2, and SERCA2a and shows that such therapeutic benefit is mediated by a more effective recovery of gene network regulation.
Collapse
Affiliation(s)
| | - Florence Tatin
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Fransky Hantelys
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Benoît Lebas
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Denis Calise
- UMS 006, Université de Toulouse, INSERM, 31432 Toulouse, France
| | - Oksana Kunduzova
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Bernard Masri
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Françoise Pujol
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Pierre Sicard
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Philippe Valet
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Jérôme Roncalli
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Xavier Chaufour
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Barbara Garmy-Susini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Angelo Parini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Anne-Catherine Prats
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France.
| |
Collapse
|
45
|
Guo W, Diao Z, Liu W. Asymmetric dimethylarginine downregulates sarco/endoplasmic reticulum calcium‑ATPase 3 and induces endoplasmic reticulum stress in human umbilical vein endothelial cells. Mol Med Rep 2017; 16:7541-7547. [PMID: 28944875 PMCID: PMC5865888 DOI: 10.3892/mmr.2017.7529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 09/06/2017] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of mortality in patients with chronic kidney disease. Endothelial cell injury and apoptosis may promote atherosclerosis and cardiovascular disease. The present study investigated the potential mechanisms of asymmetric dimethylarginine (ADMA)‑induced apoptosis in human umbilical vein endothelial cells (HUVECs). It was demonstrated that ADMA decreased B‑cell lymphoma‑2 expression and increased cleaved‑caspase‑3 expression. Furthermore, terminal deoxynucleotidyl transferase (TdT)‑mediated‑digoxigenin‑11‑dUTP nick end labeling results indicated that ADMA induced apoptosis in HUVECs. These results suggest a potential mechanism of ADMA‑induced endothelial cell injury. It was also verified that ADMA induced the expression of phosphorylated protein kinase RNA‑like ER kinase, inositol requiring enzyme‑1, C/EBP homologous protein and glucose‑regulated protein, indicating activation of the endoplasmic reticulum (ER) stress response. Impaired function of sarco/endoplasmic reticulum calcium‑ATPase (SERCA) is considered a major contributor to ER stress. It was demonstrated that ADMA induced a significant downregulation of SERCA3, however not SERCA2b. Overall, the results indicated that ADMA induced apoptosis in HUVECs, and that this effect was closely associated with induction of ER stress and decreased SERCA3 expression.
Collapse
Affiliation(s)
- Weikang Guo
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, P.R. China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, P.R. China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
46
|
Tapia-Vieyra JV, Delgado-Coello B, Mas-Oliva J. Atherosclerosis and Cancer; A Resemblance with Far-reaching Implications. Arch Med Res 2017; 48:12-26. [PMID: 28577865 DOI: 10.1016/j.arcmed.2017.03.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/02/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis and cancer are chronic diseases considered two of the main causes of death all over the world. Taking into account that both diseases are multifactorial, they share not only several important molecular pathways but also many ethiological and mechanistical processes from the very early stages of development up to the advanced forms in both pathologies. Factors involved in their progression comprise genetic alterations, inflammatory processes, uncontrolled cell proliferation and oxidative stress, as the most important ones. The fact that external effectors such as an infective process or a chemical insult have been proposed to initiate the transformation of cells in the artery wall and the process of atherogenesis, emphasizes many similarities with the progression of the neoplastic process in cancer. Deregulation of cell proliferation and therefore cell cycle progression, changes in the synthesis of important transcription factors as well as adhesion molecules, an alteration in the control of angiogenesis and the molecular similarities that follow chronic inflammation, are just a few of the processes that become part of the phenomena that closely correlates atherosclerosis and cancer. The aim of the present study is therefore, to provide new evidence as well as to discuss new approaches that might promote the identification of closer molecular ties between these two pathologies that would permit the recognition of atherosclerosis as a pathological process with a very close resemblance to the way a neoplastic process develops, that might eventually lead to novel ways of treatment.
Collapse
Affiliation(s)
| | - Blanca Delgado-Coello
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
47
|
Jardín I, López JJ, Diez R, Sánchez-Collado J, Cantonero C, Albarrán L, Woodard GE, Redondo PC, Salido GM, Smani T, Rosado JA. TRPs in Pain Sensation. Front Physiol 2017. [PMID: 28649203 PMCID: PMC5465271 DOI: 10.3389/fphys.2017.00392] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
According to the International Association for the Study of Pain (IASP) pain is characterized as an "unpleasant sensory and emotional experience associated with actual or potential tissue damage". The TRP super-family, compressing up to 28 isoforms in mammals, mediates a myriad of physiological and pathophysiological processes, pain among them. TRP channel might be constituted by similar or different TRP subunits, which will result in the formation of homomeric or heteromeric channels with distinct properties and functions. In this review we will discuss about the function of TRPs in pain, focusing on TRP channles that participate in the transduction of noxious sensation, especially TRPV1 and TRPA1, their expression in nociceptors and their sensitivity to a large number of physical and chemical stimuli.
Collapse
Affiliation(s)
- Isaac Jardín
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José J López
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Raquel Diez
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José Sánchez-Collado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Carlos Cantonero
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Letizia Albarrán
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health SciencesBethesda, MD, United States
| | - Pedro C Redondo
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Ginés M Salido
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Sevilla, University of SevilleSevilla, Spain
| | - Juan A Rosado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| |
Collapse
|
48
|
Histopathological and Immunological Characteristics of Tachycardia-Induced Cardiomyopathy. J Am Coll Cardiol 2017; 69:2160-2172. [DOI: 10.1016/j.jacc.2017.02.049] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 12/17/2022]
|
49
|
Grossi M, Bhattachariya A, Nordström I, Turczyńska KM, Svensson D, Albinsson S, Nilsson BO, Hellstrand P. Pyk2 inhibition promotes contractile differentiation in arterial smooth muscle. J Cell Physiol 2017; 232:3088-3102. [PMID: 28019664 DOI: 10.1002/jcp.25760] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 01/12/2023]
Abstract
Modulation from contractile to synthetic phenotype of vascular smooth muscle cells is a central process in disorders involving compromised integrity of the vascular wall. Phenotype modulation has been shown to include transition from voltage-dependent toward voltage-independent regulation of the intracellular calcium level, and inhibition of non-voltage dependent calcium influx contributes to maintenance of the contractile phenotype. One possible mediator of calcium-dependent signaling is the FAK-family non-receptor protein kinase Pyk2, which is activated by a number of stimuli in a calcium-dependent manner. We used the Pyk2 inhibitor PF-4594755 and Pyk2 siRNA to investigate the role of Pyk2 in phenotype modulation in rat carotid artery smooth muscle cells and in cultured intact arteries. Pyk2 inhibition promoted the expression of smooth muscle markers at the mRNA and protein levels under stimulation by FBS or PDGF-BB and counteracted phenotype shift in cultured intact carotid arteries and balloon injury ex vivo. During long-term (24-96 hr) treatment with PF-4594755, smooth muscle markers increased before cell proliferation was inhibited, correlating with decreased KLF4 expression and differing from effects of MEK inhibition. The Pyk2 inhibitor reduced Orai1 and preserved SERCA2a expression in carotid artery segments in organ culture, and eliminated the inhibitory effect of PDGF stimulation on L-type calcium channel and large-conductance calcium-activated potassium channel expression in carotid cells. Basal intracellular calcium level, calcium wave activity, and store-operated calcium influx were reduced after Pyk2 inhibition of growth-stimulated cells. Pyk2 inhibition may provide an interesting approach for preserving vascular smooth muscle differentiation under pathophysiological conditions.
Collapse
Affiliation(s)
- Mario Grossi
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Ina Nordström
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Daniel Svensson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
50
|
Regulation of Calcium Homeostasis by ER Redox: A Close-Up of the ER/Mitochondria Connection. J Mol Biol 2017; 429:620-632. [PMID: 28137421 DOI: 10.1016/j.jmb.2017.01.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 01/17/2023]
Abstract
Calcium signaling plays an important role in cell survival by influencing mitochondria-related processes such as energy production and apoptosis. The endoplasmic reticulum (ER) is the main storage compartment for cell calcium (Ca2+; ~60-500μM), and the Ca2+ released by the ER has a prompt effect on the homeostasis of the juxtaposed mitochondria. Recent findings have highlighted a close connection between ER redox and Ca2+ signaling that is mediated by Ca2+-handling proteins. This paper describes the redox-regulated mediators and mechanisms that orchestrate Ca2+ signals from the ER to mitochondria.
Collapse
|