1
|
Gomaa AAE, Zeid AMA, Nagy IM, Zahran AM. The effect of genetic polymorphisms in STIM1 and ORAI1 on erythropoietin resistance in Egyptian patients with end-stage renal disease. Clin Chim Acta 2025; 564:119948. [PMID: 39214396 DOI: 10.1016/j.cca.2024.119948] [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: 08/02/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic renal failure (CRF) is an incurable disease with unique challenges. Anemia is a frequent complication affecting dialysis patients. Erythropoietin (EPO) is used to treat anemia, but a poor response may result. We investigated genetic polymorphisms of store-operated calcium channel (SOC) signaling, an important erythropoietin-activated pathway that may induce EPO resistance in patients with renal failure. A total of 108 end stage renal disease (ESRD) patients were selected for this study. Patients were divided into two groups according to their erythropoietin resistance index (ERI): 39 patients with an ERI>10 and 69 patients with an ERI<10. We selected four tagging single nucleotide polymorphisms (tSNPs) in STIM1 and five in ORAI1 in our study. A polymerase chain reaction was performed, and genotyping against EPO resistance was correlated. Patients with the AG genotype of rs1561876 in STIM1, the TC genotype of rs6486795 in ORAI1, and the TG or GG genotypes of rs12320939 in ORAI1 were associated with an increased risk of erythropoietin resistance. Overall, we reported a moderately significant relationship between genetic polymorphisms of STIM1 and EPO resistance. We also reported a highly significant relationship between genetic polymorphisms of ORAI1 and EPO resistance. The (A-A-G) haplotype of STIM1 and the (G-T-G-T-A, G-C-G-C-G, or G-T-T-C-G) haplotypes of ORAI1 were significantly associated with EPO resistance.
Collapse
Affiliation(s)
- Azza A E Gomaa
- Internal Medicine Department, Menofia University, Menofia, Egypt.
| | - Amany M A Zeid
- Clinical Pathology Department, Menofia University, Menofia, Egypt
| | - Ibrahim M Nagy
- Medicinal Chemistry Department, Menofia University, Menofia, Egypt.
| | - Ahmed M Zahran
- Internal Medicine Department, Menofia University, Menofia, Egypt
| |
Collapse
|
2
|
Cacheux M, Velasco J, Wu X, Strauss B, Akar FG. Cardiomyocyte STIM1 downregulation exacerbates post-Myocardial Infarction remodeling by dysregulating mitochondrial ultrastructure and metabolic signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.625436. [PMID: 39713399 PMCID: PMC11661096 DOI: 10.1101/2024.12.05.625436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background Loss of stromal interaction molecule 1 (STIM1) expression in smooth muscle cells protects against ischemia-reperfusion (I/R) injury. Whether and how decreased STIM1 expression in cardiomyocytes (CM) impacts cardiac remodeling in response to I/R injury remains unknown. Objective To examine mechanisms by which decreased CM-STIM1 expression in the adult heart modulates cardiac function before and after I/R injury. Methods 8-week old mice underwent cardiotropic AAV9-mediated gene transfer of shRNA directed against STIM1 (shSTIM1). Control (Ctrl) mice underwent shRNA luciferase or PBS injections. Ctrl and shSTIM1 mice were then challenged by 30-min coronary occlusion to induce MI, in-vivo . Mechanical, structural and electrophysiological (EP) properties were compared 1-week following MI. In a second cohort of mice, the impact of CM-STIM1 knockdown per se on upstream metabolic signaling, mitochondrial ultrastructure, and electrophysiological properties were studied. Results CM-STIM1 expression was markedly decreased in shSTIM1 vs Ctrl hearts. Challenge with in-vivo I/R injury resulted in more pronounced (p<0.0001) LV dysfunction indexed by % drop in fractional shortening in shSTIM1 (44.3%) vs Ctrl (12.2%) hearts 1-week post-MI. Similarly, post-MI structural remodeling and the extent of fibrosis were more severe in shSTIM1 vs Ctrl despite comparable infarct size (p=0.514). Consistently, shSTIM1 exhibited greater impairment in post-MI EP function including predisposition to spatially-discordant action potential alternans. To understand mechanisms underlying this differential remodeling, we examined the impact of CM-STIM1 downregulation on mitochondrial ultrastructure and regulation by metabolic signaling. Quantification of mitochondrial morphology revealed smaller, more rounded mitochondria caused by CM-STIM1 downregulation per se . Underlying these changes was a marked (by 55%, p=0.0057) increase in phosphorylated (p)DRP1 at S616 along with reduced OPA1 expression. Mitochondrial alterations were associated with significant decreases in AMPK downstream signaling with loss of phosphorylated-to-total Raptor and ACC expression in shSTIM1-vs-Ctrl hearts consistent with impaired fatty acid oxidation. These MI-independent metabolic alterations coincided with higher pro-arrhythmic vulnerability under conditions of elevated heart rate. Conclusions Our findings reveal that decreased CM-STIM1 expression exacerbates post-MI remodeling likely by altering metabolic processes and mitochondrial network dynamics.Functionally, STIM1-dependent mitochondrial alterations impact EP function during conditions of elevated heart rate even without the confounding influence of MI.
Collapse
|
3
|
Borge SJ, Sennels HP, Schwarz P, Jørgensen HL. Diurnal fluctuations in biochemical parameters related to calcium homeostasis - the Bispebjerg study of diurnal variations. Scand J Clin Lab Invest 2024; 84:305-310. [PMID: 39163206 DOI: 10.1080/00365513.2024.2392116] [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: 04/11/2024] [Revised: 06/21/2024] [Accepted: 08/10/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE This aim of this study was to assess the possible association between diurnal oscillations and biochemical markers associated with calcium homeostasis. This included the markers parathyroid hormone (PTH), total calcium, total alkaline phosphatase, phosphate, and 25-hydroxyvitamin D (25-OH-D). By examining the influence of circadian rhythms on these parameters, the study aimed to deepen the understanding of calcium metabolism dynamics and its clinical implications. PATIENTS AND METHODS Blood samples from 24 Caucasian male volunteers aged 20 to 40 (mean age 26) with normal pulse, blood pressure, and BMI were analyzed for biochemical markers related to calcium homeostasis. Data was obtained from the Bispebjerg study of diurnal variations. Blood samples were collected every three hours over a 24-hour period. Patients were fasting from 22:00 to 09:00. The participants spent 24 h in the hospital ward, receiving regular meals and engaging in low-intensity activities. They experienced 15 h of daylight and 9 h of complete darkness during sleep. Diurnal oscillations were analyzed using cosinor analysis with statistical significance set at p < 0.05. RESULTS Total calcium, phosphate, and PTH exhibited significant diurnal variations. Total calcium and PTH were inversely synchronized while PTH and phosphate oscillated in synchronization. The three parameters showed relatively large amplitude/reference range ratios from 25.4% to 41.5%. CONCLUSION This study found notable fluctuations in total calcium, phosphate, and PTH levels over a 24-hour cycle, while 25-OH-D and total alkaline phosphatase remained consistent. It highlights the importance of considering sampling times for total calcium, PTH, and phosphate in clinical settings.
Collapse
Affiliation(s)
- Silje J Borge
- Department of Clinical Biochemistry, Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Henriette P Sennels
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, København, NV, Denmark
| | - Peter Schwarz
- Department of Endocrinology, Rigshospitalet, København Ø, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henrik L Jørgensen
- Department of Clinical Biochemistry, Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Lee L, Yoast R, Emrich S, Trebak M, Kirk V, Sneyd J. Emergence of broad cytosolic Ca 2+ oscillations in the absence of CRAC channels: A model for CRAC-mediated negative feedback on PLC and Ca 2+ oscillations through PKC. J Theor Biol 2024; 581:111740. [PMID: 38253220 DOI: 10.1016/j.jtbi.2024.111740] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
The role of Ca2+ release-activated Ca2+ (CRAC) channels mediated by ORAI isoforms in calcium signalling has been extensively investigated. It has been shown that the presence or absence of different isoforms has a significant effect on store-operated calcium entry (SOCE). Yoast et al. (2020) showed that, in addition to the reported narrow-spike oscillations (whereby cytosolic calcium decreases quickly after a sharp increase), ORAI1 knockout HEK293 cells were able to oscillate with broad-spike oscillations (whereby cytosolic calcium decreases in a prolonged manner after a sharp increase) when stimulated with a muscarinic agonist. This suggests that Ca2+ influx through ORAI-mediated CRAC channels negatively regulates the duration of Ca2+ oscillations. We hypothesise that, through the activation of protein kinase C (PKC), ORAI1 negatively regulates phospholipase C (PLC) activity to decrease inositol 1,4,5-trisphosphate (IP3) production and limit the duration of agonist-evoked Ca2+ oscillations. Based on this hypothesis, we construct a new mathematical model, which shows that the formation of broad-spike oscillations is highly dependent on the absence of ORAI1. Predictions of this model are consistent with the experimental results.
Collapse
Affiliation(s)
- Lloyd Lee
- Department of Mathematics, University of Auckland, 1142 Auckland, New Zealand.
| | - Ryan Yoast
- Graduate Program in Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Scott Emrich
- Graduate Program in Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA
| | - Vivien Kirk
- Department of Mathematics, University of Auckland, 1142 Auckland, New Zealand
| | - James Sneyd
- Department of Mathematics, University of Auckland, 1142 Auckland, New Zealand
| |
Collapse
|
5
|
Hermes J, Borisova V, Kockskämper J. Store-Operated Calcium Entry Increases Nuclear Calcium in Adult Rat Atrial and Ventricular Cardiomyocytes. Cells 2023; 12:2690. [PMID: 38067118 PMCID: PMC10705675 DOI: 10.3390/cells12232690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Store-operated calcium entry (SOCE) in cardiomyocytes may be involved in cardiac remodeling, but the underlying mechanisms remain elusive. We hypothesized that SOCE may increase nuclear calcium, which alters gene expression via calcium/calmodulin-dependent enzyme signaling, and elucidated the underlying cellular mechanisms. An experimental protocol was established in isolated adult rat cardiomyocytes to elicit SOCE by re-addition of calcium following complete depletion of sarcoplasmic reticulum (SR) calcium and to quantify SOCE in relation to the electrically stimulated calcium transient (CaT) measured in the same cell before SR depletion. Using confocal imaging, calcium changes were recorded simultaneously in the cytosol and in the nucleus of the cell. In ventricular myocytes, SOCE was observed in the cytosol and nucleus amounting to ≈15% and ≈25% of the respective CaT. There was a linear correlation between the SOCE-mediated calcium increase in the cytosol and nucleus. Inhibitors of TRPC or Orai channels reduced SOCE by ≈33-67%, whereas detubulation did not. In atrial myocytes, SOCE with similar characteristics was observed in the cytosol and nucleus. However, the SOCE amplitudes in atrial myocytes were ≈two-fold larger than in ventricular myocytes, and this was associated with ≈1.4- to 3.6-fold larger expression of putative SOCE proteins (TRPC1, 3, 6, and STIM1) in atrial tissue. The results indicated that SOCE in atrial and ventricular myocytes is able to cause robust calcium increases in the nucleus and that both TRPC and Orai channels may contribute to SOCE in adult cardiomyocytes.
Collapse
Affiliation(s)
- Julia Hermes
- Institute for Pharmacology and Clinical Pharmacy, Biochemical and Pharmacological Centre (BPC) Marburg, University of Marburg, Karl-von-Frisch-Str. 2 K|03, 35043 Marburg, Germany
| | - Vesela Borisova
- Institute for Pharmacology and Clinical Pharmacy, Biochemical and Pharmacological Centre (BPC) Marburg, University of Marburg, Karl-von-Frisch-Str. 2 K|03, 35043 Marburg, Germany
- Department of Pharmacology and Clinical Pharmacology and Therapeutics, Medical University of Varna, Varna 9002, 55 Marin Drinov str., Bulgaria
| | - Jens Kockskämper
- Institute for Pharmacology and Clinical Pharmacy, Biochemical and Pharmacological Centre (BPC) Marburg, University of Marburg, Karl-von-Frisch-Str. 2 K|03, 35043 Marburg, Germany
| |
Collapse
|
6
|
Li HX, Ma Y, Yan YX, Zhai XK, Xin MY, Wang T, Xu DC, Song YT, Song CD, Pan CX. The purified extract of steamed Panax ginseng protects cardiomyocyte from ischemic injury via caveolin-1 phosphorylation-mediating calcium influx. J Ginseng Res 2023; 47:755-765. [PMID: 38107394 PMCID: PMC10721475 DOI: 10.1016/j.jgr.2023.07.003] [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: 09/01/2022] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 12/19/2023] Open
Abstract
Background Caveolin-1, the scaffolding protein of cholesterol-rich invaginations, plays an important role in store-operated Ca2+ influx and its phosphorylation at Tyr14 (p-caveolin-1) is vital to mobilize protection against myocardial ischemia (MI) injury. SOCE, comprising STIM1, ORAI1 and TRPC1, contributes to intracellular Ca2+ ([Ca2+]i) accumulation in cardiomyocytes. The purified extract of steamed Panax ginseng (EPG) attenuated [Ca2+]i overload against MI injury. Thus, the aim of this study was to investigate the possibility of EPG affecting p-caveolin-1 to further mediate SOCE/[Ca2+]i against MI injury in neonatal rat cardiomyocytes and a rat model. Methods PP2, an inhibitor of p-caveolin-1, was used. Cell viability, [Ca2+]i concentration were analyzed in cardiomyocytes. In rats, myocardial infarct size, pathological damages, apoptosis and cardiac fibrosis were evaluated, p-caveolin-1 and STIM1 were detected by immunofluorescence, and the levels of caveolin-1, STIM1, ORAI1 and TRPC1 were determined by RT-PCR and Western blot. And, release of LDH, cTnI and BNP was measured. Results EPG, ginsenosides accounting for 57.96%, suppressed release of LDH, cTnI and BNP, and protected cardiomyocytes by inhibiting Ca2+ influx. And, EPG significantly relieved myocardial infarct size, cardiac apoptosis, fibrosis, and ultrastructure abnormality. Moreover, EPG negatively regulated SOCE via increasing p-caveolin-1 protein, decreasing ORAI1 mRNA and protein levels of ORAI1, TRPC1 and STIM1. More importantly, inhibition of the p-caveolin-1 significantly suppressed all of the above cardioprotection of EPG. Conclusions Caveolin-1 phosphorylation is involved in the protective effects of EPG against MI injury via increasing p-caveolin-1 to negatively regulate SOCE/[Ca2+]i.
Collapse
Affiliation(s)
- Hai-Xia Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, China
| | - Yan Ma
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Yu-Xiao Yan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Xin-Ke Zhai
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Meng-Yu Xin
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Tian Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Dong-Cao Xu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Yu-Tong Song
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Chun-Dong Song
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, 9 Renmin Road, Zhengzhou, Henan Province, China
| | - Cheng-Xue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| |
Collapse
|
7
|
Liu P, Yang Z, Wang Y, Sun A. Role of STIM1 in the Regulation of Cardiac Energy Substrate Preference. Int J Mol Sci 2023; 24:13188. [PMID: 37685995 PMCID: PMC10487555 DOI: 10.3390/ijms241713188] [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: 07/15/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The heart requires a variety of energy substrates to maintain proper contractile function. Glucose and long-chain fatty acids (FA) are the major cardiac metabolic substrates under physiological conditions. Upon stress, a shift of cardiac substrate preference toward either glucose or FA is associated with cardiac diseases. For example, in pressure-overloaded hypertrophic hearts, there is a long-lasting substrate shift toward glucose, while in hearts with diabetic cardiomyopathy, the fuel is switched toward FA. Stromal interaction molecule 1 (STIM1), a well-established calcium (Ca2+) sensor of endoplasmic reticulum (ER) Ca2+ store, is increasingly recognized as a critical player in mediating both cardiac hypertrophy and diabetic cardiomyopathy. However, the cause-effect relationship between STIM1 and glucose/FA metabolism and the possible mechanisms by which STIM1 is involved in these cardiac metabolic diseases are poorly understood. In this review, we first discussed STIM1-dependent signaling in cardiomyocytes and metabolic changes in cardiac hypertrophy and diabetic cardiomyopathy. Second, we provided examples of the involvement of STIM1 in energy metabolism to discuss the emerging role of STIM1 in the regulation of energy substrate preference in metabolic cardiac diseases and speculated the corresponding underlying molecular mechanisms of the crosstalk between STIM1 and cardiac energy substrate preference. Finally, we briefly discussed and presented future perspectives on the possibility of targeting STIM1 to rescue cardiac metabolic diseases. Taken together, STIM1 emerges as a key player in regulating cardiac energy substrate preference, and revealing the underlying molecular mechanisms by which STIM1 mediates cardiac energy metabolism could be helpful to find novel targets to prevent or treat cardiac metabolic diseases.
Collapse
Affiliation(s)
- Panpan Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Zhuli Yang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Aomin Sun
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
8
|
Zhang W, Sun Y, Yang Y, Chen Y. Impaired intracellular calcium homeostasis enhances protein O-GlcNAcylation and promotes vascular calcification and stiffness in diabetes. Redox Biol 2023; 63:102720. [PMID: 37230005 PMCID: PMC10225928 DOI: 10.1016/j.redox.2023.102720] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/13/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Vascular calcification is accelerated in patients with diabetes mellitus and increases risk of cardiovascular events and mortality. Vascular smooth muscle cells (VSMC) play a key role in regulating vascular tone and contribute significantly to the development of diabetic vasculopathy. In this study, the function of stromal interaction molecule 1 (STIM1), an important regulator for intracellular calcium homeostasis, in diabetic vascular calcification was investigated, and the underlying molecular mechanisms were uncovered. A SMC-specific STIM1 deletion mouse model (STIM1Δ/Δ) was generated by breeding the STIM1 floxed mice (STIM1f/f) with SM22α-Cre transgenic mice. Using aortic arteries from the STIM1Δ/Δ mice and their STIM1f/f littermates, we found that SMC-specific STIM1 deletion induced calcification of aortic arteries cultured in osteogenic media ex vivo. Furthermore, STIM1 deficiency promoted osteogenic differentiation and calcification of VSMC from the STIM1Δ/Δ mice. In the low-dose streptozotocin (STZ)-induced mouse model of diabetes, SMC-specific STIM1 deletion markedly enhanced STZ-induced vascular calcification and stiffness in the STIM1Δ/Δ mice. The diabetic mice with SMC-specific STIM1 ablation also exhibited increased aortic expression of the key osteogenic transcription factor, Runx2, and protein O-GlcNAcylation, an important post-translational modulation that we have reported to promote vascular calcification and stiffness in diabetes. Consistently, elevation of O-GlcNAcylation was demonstrated in aortic arteries and VSMC from the STIM1Δ/Δ mice. Inhibition of O-GlcNAcylation with a pharmacological inhibitor abolished STIM1 deficiency-induced VSMC calcification, supporting a critical role of O-GlcNAcylation in mediating STIM1 deficiency-induced VSMC calcification. Mechanistically, we identified that STIM1 deficiency resulted in impaired calcium homeostasis, which activated calcium signaling and increased endoplasmic reticulum (ER) stress in VSMC, while inhibition of ER stress attenuated STIM1-induced elevation of protein O-GlcNAcylation. In conclusion, the study has demonstrated a causative role of SMC-expressed STIM1 in regulating vascular calcification and stiffness in diabetes. We have further identified a novel mechanisms underlying STIM1 deficiency-induced impairment of calcium homeostasis and ER stress in upregulation of protein O-GlcNAcylation in VSMC, which promotes VSMC osteogenic differentiation and calcification in diabetes.
Collapse
Affiliation(s)
- Weiping Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Cardiology, First Affiliated Hospital of Xi'An JiaoTong University, Xi'An, PR China
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Youfeng Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
9
|
Bonilla IM, Baine S, Pokrass A, Mariángelo JIE, Kalyanasundaram A, Bogdanov V, Mezache L, Sakuta G, Beard CM, Belevych A, Tikunova S, Terentyeva R, Terentyev D, Davis J, Veeraraghavan R, Carnes CA, Györke S. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts. J Appl Physiol (1985) 2023; 134:1287-1299. [PMID: 36995910 PMCID: PMC10190841 DOI: 10.1152/japplphysiol.00363.2022] [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: 07/05/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Cardiac stromal interaction molecule 1 (STIM1), a key mediator of store-operated Ca2+ entry (SOCE), is a known determinant of cardiomyocyte pathological growth in hypertrophic cardiomyopathy. We examined the role of STIM1 and SOCE in response to exercise-dependent physiological hypertrophy. Wild-type (WT) mice subjected to exercise training (WT-Ex) showed a significant increase in exercise capacity and heart weight compared with sedentary (WT-Sed) mice. Moreover, myocytes from WT-Ex hearts displayed an increase in length, but not width, compared with WT-Sed myocytes. Conversely, exercised cardiac-specific STIM1 knock-out mice (cSTIM1KO-Ex), although displaying significant increase in heart weight and cardiac dilation, evidenced no changes in myocyte size and displayed a decreased exercise capacity, impaired cardiac function, and premature death compared with sedentary cardiac-specific STIM1 knock-out mice (cSTIM1KO-Sed). Confocal Ca2+ imaging demonstrated enhanced SOCE in WT-Ex myocytes compared with WT-Sed myocytes with no measurable SOCE detected in cSTIM1KO myocytes. Exercise training induced a significant increase in cardiac phospho-Akt Ser473 in WT mice but not in cSTIM1KO mice. No differences were observed in phosphorylation of mammalian target of rapamycin (mTOR) and glycogen synthase kinase (GSK) in exercised versus sedentary cSTIM1KO mice hearts. cSTIM1KO-Sed mice showed increased basal MAPK phosphorylation compared with WT-Sed that was not altered by exercise training. Finally, histological analysis revealed exercise resulted in increased autophagy in cSTIM1KO but not in WT myocytes. Taken together, our results suggest that adaptive cardiac hypertrophy in response to exercise training involves STIM1-mediated SOCE. Our results demonstrate that STIM1 is involved in and essential for the myocyte longitudinal growth and mTOR activation in response to endurance exercise training.NEW & NOTEWORTHY Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, United States
| | - Stephen Baine
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Anastasia Pokrass
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Juan Ignacio Elio Mariángelo
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart & Lung Research Institute, Columbus, Ohio, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Galina Sakuta
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Casey M Beard
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Andriy Belevych
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Jonathan Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Cynthia A Carnes
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Sandor Györke
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
10
|
Lim S, Oh S, Nguyen QTN, Kim M, Zheng S, Fang M, Yi TH. Rosa davurica Inhibited Allergic Mediators by Regulating Calcium and Histamine Signaling Pathways. PLANTS (BASEL, SWITZERLAND) 2023; 12:1572. [PMID: 37050198 PMCID: PMC10097250 DOI: 10.3390/plants12071572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 06/19/2023]
Abstract
Rosa davurica Pall. exhibits antioxidant, antiviral, and anti-inflammatory properties; however, its pharmacological mechanism in allergy is yet to be understood. This study confirmed the effects of R. davurica Pall. leaf extract (RLE) on allergy as a new promising material. To evaluate the therapeutic potential of RLE against allergy, we investigated the effects of RLE on the regulatory β-hexosaminidase, histamine, histidine decarboxylase (HDC), Ca2+ influx, nitric oxide (NO), and cytokines induced by lipopolysaccharide (LPS) and DNP-IgE/BSA in Raw 264.7 and RBL-2H3 cells. Furthermore, we examined the effects of RLE on the signaling pathways of mitogen-activated protein kinase (MAPK) and Ca2+ pathways. After stimulating Raw 264.7 cells with LPS, RLE reduced the release of inflammatory mediators, such as NO, cyclooxygenase (COX)-2, inducible nitric oxygen synthase (iNOS), interleukin (IL)-1β, -6, and tumor necrosis factor (TNF)-α. Also, RLE reduced the β-hexosaminidase, histamine, HDC, Ca2+ influx, Ca2+ pathways, and phosphorylation of MAPK in DNP-IgE/BSA-stimulated RBL-2H3 cells. Our studies indicated that RLE is a valuable ingredient for treating allergic diseases by regulating cytokine release from macrophages and mast cell degranulation. Consequently, these results suggested that RLE may serve as a possible alternative promising material for treating allergies.
Collapse
Affiliation(s)
- Seojun Lim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (S.L.); (Q.T.N.N.); (M.K.); (M.F.)
| | - Sarang Oh
- Snowwhitefactory Co., Ltd., 807 Nonhyeon-ro, Gangnam-gu, Seoul 06032, Republic of Korea; (S.O.); (S.Z.)
| | - Quynh T. N. Nguyen
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (S.L.); (Q.T.N.N.); (M.K.); (M.F.)
| | - Myeongju Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (S.L.); (Q.T.N.N.); (M.K.); (M.F.)
| | - Shengdao Zheng
- Snowwhitefactory Co., Ltd., 807 Nonhyeon-ro, Gangnam-gu, Seoul 06032, Republic of Korea; (S.O.); (S.Z.)
| | - Minzhe Fang
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (S.L.); (Q.T.N.N.); (M.K.); (M.F.)
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (S.L.); (Q.T.N.N.); (M.K.); (M.F.)
| |
Collapse
|
11
|
Xiao H, Lu H, Xue Y, Jia Z, Dai M, He K, Zhao R. Deleterious effect in endothelin receptor-mediated coronary artery smooth muscle contractility in high-salt diet rats. Nutr Metab Cardiovasc Dis 2023; 33:234-244. [PMID: 36404239 DOI: 10.1016/j.numecd.2022.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS High-salt diet has been suggested to increase the risk of heart disease. However, the mechanisms underlying coronary artery tension dysfunction caused by high-salt diet are unclear. Previous studies have shown that coronary artery spasm is often induced by endothelin-1 (ET-1) and thromboxane, leading to myocardial ischemia, while the store-operated Ca2+ entry (SOCE) function of coronary smooth muscle is very important in this process. METHODS AND RESULTS Tension measurements of endothelium-denuded coronary artery ring segments showed that vasocontraction induced by U46619, ET-1, orSTIM1/Orai1-mediated SOCE was significantly lower in 4% high-salt diet rats than in control rats fed a regular diet. The results of western blotting and immunohistochemistry assays showed lower expression levels of endothelial receptors ETA and ETB, STIM1 and Orai1 in coronary artery of high-salt intake rats compared with control rats. Fibrosis was observed by using Masson's trichrome staining and picrosirius red staining. The plasma ET-1 concentration in high-salt diet rats was significantly higher than that of controls. The interventricular septum and posterior wall of high-salt diet rats were significantly thickened. CONCLUSION Our findings indicated that coronary artery tension was significantly decreased in 4% high-salt diet rats and that this decrease may be due to the change of endothelin receptor and its downstream pathway SOCE related protein expression in coronary artery. Coronary fibrosis was observed in rats fed with high-salt diet. This study provides potential mechanistic insights into high-salt intake-induced heart disease.
Collapse
Affiliation(s)
- Hui Xiao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Haoyang Lu
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yangcheng Xue
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Zhuoran Jia
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Manyu Dai
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ke He
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| | - Ren Zhao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| |
Collapse
|
12
|
β-carotene alleviates LPS-induced inflammation through regulating STIM1/ORAI1 expression in bovine mammary epithelial cells. Int Immunopharmacol 2022; 113:109377. [DOI: 10.1016/j.intimp.2022.109377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
|
13
|
Komorowska D, Radzik T, Kalenik S, Rodacka A. Natural Radiosensitizers in Radiotherapy: Cancer Treatment by Combining Ionizing Radiation with Resveratrol. Int J Mol Sci 2022; 23:ijms231810627. [PMID: 36142554 PMCID: PMC9501384 DOI: 10.3390/ijms231810627] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional cancer treatment is mainly based on the surgical removal of the tumor followed by radiotherapy and/or chemotherapy. When surgical removal is not possible, radiotherapy and, less often, chemotherapy is the only way to treat patients. However, despite significant progress in understanding the molecular mechanisms of carcinogenesis and developments in modern radiotherapy techniques, radiotherapy (alone or in combination) does not always guarantee treatment success. One of the main causes is the radioresistance of cancer cells. Increasing the radiosensitivity of cancer cells improves the processes leading to their elimination during radiotherapy and prolonging the survival of cancer patients. In order to enhance the effect of radiotherapy in the treatment of radioresistant neoplasms, radiosensitizers are used. In clinical practice, synthetic radiosensitizers are commonly applied, but scientists have recently focused on using natural products (phytocompounds) as adjuvants in radiotherapy. In this review article, we only discuss naturally occurring radiosensitizers currently in clinical trials (paclitaxel, curcumin, genistein, and papaverine) and those whose radiation sensitizing effects, such as resveratrol, have been repeatedly confirmed by many independent studies.
Collapse
Affiliation(s)
- Dominika Komorowska
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | - Tomasz Radzik
- MARINEX International, 4 Placowa St., 93-446 Lodz, Poland
| | - Sebastian Kalenik
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | - Aleksandra Rodacka
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
- Correspondence: ; Fax: +48-426354473
| |
Collapse
|
14
|
Ye J, Li M, Li Q, Jia Z, Hu X, Zhao G, Zhi S, Hong G, Lu Z. Activation of STIM1/Orai1‑mediated SOCE in sepsis‑induced myocardial depression. Mol Med Rep 2022; 26:259. [PMID: 35713214 DOI: 10.3892/mmr.2022.12775] [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: 02/04/2021] [Accepted: 02/21/2022] [Indexed: 11/05/2022] Open
Abstract
Unbalanced Ca2+ homeostasis serves an essential role in the occurrence and development of septic myocardial injury. However, the mechanism of Ca2+ homeostasis in septic myocardial depression is poorly understood due to the complexity of Ca2+ transporters in excitable cells. It was therefore hypothesized that cardiac dysfunction, myocardial injury and cardiac apoptosis in septic myocardial depression are associated with elevated intracellular Ca2+ concentrations caused by stromal interaction molecule 1 (STIM1)/Orai calcium release‑activated calcium modulator 1 (Orai1)‑mediated store‑operated Ca2+ entry (SOCE). A septic myocardial depression model was established using the cecal ligation and puncture operation (CLP) in mice and was simulated in H9C2 cells via lipopolysaccharide (LPS) stimulation. Cardiac function, myocardial injury, cardiac apoptosis and the expression levels of Bax, Bcl‑2, STIM1 and Orai1 were quantified in vivo at 6, 12 and 24 h. Changes in the intracellular Ca2+ concentration, SOCE and the distribution of STIM1 were assessed in vitro within 6 h. The morphological changes of heart tissue were observed by hematoxylin‑eosin staining. Myocardial cellular apoptosis was determined by TUNEL method. The expression of Bax, Bcl‑2, STIM1 and Orai1 were visualized by western blot. Cytosolic calcium concentration and SOCE were evaluated by confocal microscopy. The results demonstrated that cardiac contractile function was significantly reduced at 6 h and morphological changes in cardiac tissues, as well as the myocardial apoptosis rate, were markedly increased at 6, 12 and 24 h following CLP. mRNA and protein expression levels of Bax/Bcl‑2 were significantly enhanced at 6 and 12 h and glycosylation of Orai1 in the myocardium of septic mice was significantly increased at 6 h following CLP. The intracellular Ca2+ concentration, SOCE, was significantly increased at 1‑2 h and the clustering and distribution of STIM1 were markedly changed in H9C2 cells at 1 and 2 h. These findings suggested that myocardial dysfunction, cardiac injury and myocardial depression may be related to increased intracellular Ca2+ concentration resulting from STIM1/Orai1‑mediated SOCE, which may provide a potential method to alleviate septic myocardial depression.
Collapse
Affiliation(s)
- Jingjing Ye
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengfang Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qiao Li
- Ultrasound Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhijun Jia
- Ultrasound Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiyi Hu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangju Zhao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shaoce Zhi
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhongqiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
15
|
Wei T, Wang J, Liang R, Chen W, Chen Y, Ma M, He A, Du Y, Zhou W, Zhang Z, Zeng X, Wang C, Lu J, Guo X, Chen XW, Wang Y, Tian R, Xiao J, Lei X. Selective inhibition reveals the regulatory function of DYRK2 in protein synthesis and calcium entry. eLife 2022; 11:e77696. [PMID: 35439114 PMCID: PMC9113749 DOI: 10.7554/elife.77696] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
The dual-specificity tyrosine phosphorylation-regulated kinase DYRK2 has emerged as a critical regulator of cellular processes. We took a chemical biology approach to gain further insights into its function. We developed C17, a potent small-molecule DYRK2 inhibitor, through multiple rounds of structure-based optimization guided by several co-crystallized structures. C17 displayed an effect on DYRK2 at a single-digit nanomolar IC50 and showed outstanding selectivity for the human kinome containing 467 other human kinases. Using C17 as a chemical probe, we further performed quantitative phosphoproteomic assays and identified several novel DYRK2 targets, including eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and stromal interaction molecule 1 (STIM1). DYRK2 phosphorylated 4E-BP1 at multiple sites, and the combined treatment of C17 with AKT and MEK inhibitors showed synergistic 4E-BP1 phosphorylation suppression. The phosphorylation of STIM1 by DYRK2 substantially increased the interaction of STIM1 with the ORAI1 channel, and C17 impeded the store-operated calcium entry process. These studies collectively further expand our understanding of DYRK2 and provide a valuable tool to pinpoint its biological function.
Collapse
Affiliation(s)
- Tiantian Wei
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Jue Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Ruqi Liang
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Wendong Chen
- SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and TechnologyShenzhenChina
| | - Yilan Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Mingzhe Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - An He
- Department of Chemistry, Southern University of Science and TechnologyShenzhenChina
| | - Yifei Du
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Wenjing Zhou
- Institute of Molecular Medicine, Peking UniversityBeijingChina
| | - Zhiying Zhang
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
| | - Xin Zeng
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Chu Wang
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Jin Lu
- Peking University Institute of Hematology, People’s HospitalBeijingChina
- Collaborative Innovation Center of HematologySuzhouChina
| | - Xing Guo
- Life Sciences Institute, Zhejiang UniversityHangzhouChina
| | - Xiao-Wei Chen
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Institute of Molecular Medicine, Peking UniversityBeijingChina
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Ruijun Tian
- SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and TechnologyShenzhenChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Junyu Xiao
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
- Beijing Advanced Innovation Center for Genomics (ICG), Peking UniversityBeijingChina
| | - Xiaoguang Lei
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking UniversityBeijingChina
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
- Institute for Cancer Research, Shenzhen Bay LaboratoryShenzhenChina
| |
Collapse
|
16
|
Wu D, Yang C, Yang M, Wu Y, Mao Y, Zhou X, Wang J, Yuan Z, Wu J. Citrinin-Induced Hepatotoxicity in Mice Is Regulated by the Ca 2+/Endoplasmic Reticulum Stress Signaling Pathway. Toxins (Basel) 2022; 14:259. [PMID: 35448868 PMCID: PMC9029441 DOI: 10.3390/toxins14040259] [Citation(s) in RCA: 3] [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: 02/07/2022] [Revised: 03/23/2022] [Accepted: 04/01/2022] [Indexed: 12/26/2022] Open
Abstract
Citrinin (CTN) is a mycotoxin found in crops and agricultural products and poses a serious threat to human and animal health. The aim of this study is to investigate the hepatotoxicity of CTN in mice and analyze its mechanisms from Ca2+-dependent endoplasmic reticulum (ER) stress perspective. We showed that CTN induced histopathological damage, caused ultrastructural changes in liver cells, and induced abnormal values of biochemical laboratory tests of some liver functions in mice. Treatment with CTN could induce nitric oxide (NO), malondialdehyde (MDA), and reactive oxygen species (ROS) accumulation in mice, accompanied with losses of activities of superoxide dismutase (SOD) and catalase (CAT), levels of glutathione (GSH), and capacities of total antioxidant (T-AOC), resulting in oxidative stress in mice. Furthermore, CTN treatment significantly increased Ca2+ accumulation, upregulated protein expressions of ER stress-mediated apoptosis signal protein (glucose regulated protein 78 (GRP78/BIP), C/EBP-homologous protein (CHOP), Caspase-12, and Caspase-3), and induced hepatocyte apoptosis. These adverse effects were counteracted by 4-phenylbutyric acid (4-PBA), an ER stress inhibitor. In summary, our results showed a possible underlying molecular mechanism for CTN that induced hepatocyte apoptosis in mice by the regulation of the Ca2+/ER stress signaling pathway.
Collapse
Affiliation(s)
- Dongyi Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Chenglin Yang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Mengran Yang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - You Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Yan Mao
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Xinyan Zhou
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Ji Wang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Zhihang Yuan
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Jing Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| |
Collapse
|
17
|
Lu T, Zhang Y, Su Y, Zhou D, Xu Q. Role of store-operated Ca2+ entry in cardiovascular disease. Cell Commun Signal 2022; 20:33. [PMID: 35303866 PMCID: PMC8932232 DOI: 10.1186/s12964-022-00829-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/14/2022] [Indexed: 01/01/2023] Open
Abstract
Store-operated channels (SOCs) are highly selective Ca2+ channels that mediate Ca2+ influx in non-excitable and excitable (i.e., skeletal and cardiac muscle) cells. These channels are triggered by Ca2+ depletion of the endoplasmic reticulum and sarcoplasmic reticulum, independently of inositol 1,4,5-trisphosphate (InsP3), which is involved in cell growth, differentiation, and gene transcription. When the Ca2+ store is depleted, stromal interaction molecule1 (STIM1) as Ca2+ sensor redistributes into discrete puncta near the plasma membrane and activates the protein Ca2+ release activated Ca2+ channel protein 1 (Orai1). Accumulating evidence suggests that SOC is associated with several physiological roles in endothelial dysfunction and vascular smooth muscle proliferation that contribute to the progression of cardiovascular disease. This review mainly elaborates on the contribution of SOC in the vasculature (endothelial cells and vascular smooth muscle cells). We will further retrospect the literature implicating a critical role for these proteins in cardiovascular disease.
Collapse
Affiliation(s)
- Ting Lu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yihua Zhang
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yong Su
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Dayan Zhou
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Qiang Xu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China.
| |
Collapse
|
18
|
Zou L, Collins HE, Young ME, Zhang J, Wende AR, Darley-Usmar VM, Chatham JC. The Identification of a Novel Calcium-Dependent Link Between NAD + and Glucose Deprivation-Induced Increases in Protein O-GlcNAcylation and ER Stress. Front Mol Biosci 2021; 8:780865. [PMID: 34950703 PMCID: PMC8691773 DOI: 10.3389/fmolb.2021.780865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/22/2021] [Indexed: 01/19/2023] Open
Abstract
The modification of proteins by O-linked β-N-acetylglucosamine (O-GlcNAc) is associated with the regulation of numerous cellular processes. Despite the importance of O-GlcNAc in mediating cellular function our understanding of the mechanisms that regulate O-GlcNAc levels is limited. One factor known to regulate protein O-GlcNAc levels is nutrient availability; however, the fact that nutrient deficient states such as ischemia increase O-GlcNAc levels suggests that other factors also contribute to regulating O-GlcNAc levels. We have previously reported that in unstressed cardiomyocytes exogenous NAD+ resulted in a time and dose dependent decrease in O-GlcNAc levels. Therefore, we postulated that NAD+ and cellular O-GlcNAc levels may be coordinately regulated. Using glucose deprivation as a model system in an immortalized human ventricular cell line, we examined the influence of extracellular NAD+ on cellular O-GlcNAc levels and ER stress in the presence and absence of glucose. We found that NAD+ completely blocked the increase in O-GlcNAc induced by glucose deprivation and suppressed the activation of ER stress. The NAD+ metabolite cyclic ADP-ribose (cADPR) had similar effects on O-GlcNAc and ER stress suggesting a common underlying mechanism. cADPR is a ryanodine receptor (RyR) agonist and like caffeine, which also activates the RyR, both mimicked the effects of NAD+. SERCA inhibition, which also reduces ER/SR Ca2+ levels had similar effects to both NAD+ and cADPR on O-GlcNAc and ER stress responses to glucose deprivation. The observation that NAD+, cADPR, and caffeine all attenuated the increase in O-GlcNAc and ER stress in response to glucose deprivation, suggests a potential common mechanism, linked to ER/SR Ca2+ levels, underlying their activation. Moreover, we showed that TRPM2, a plasma membrane cation channel was necessary for the cellular responses to glucose deprivation. Collectively, these findings support a novel Ca2+-dependent mechanism underlying glucose deprivation induced increase in O-GlcNAc and ER stress.
Collapse
Affiliation(s)
- Luyun Zou
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Helen E. Collins
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Martin E. Young
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianhua Zhang
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States,Birmingham VA Medical Center, Birmingham, AL, United States
| | - Adam R. Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Victor M. Darley-Usmar
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: John C. Chatham,
| |
Collapse
|
19
|
Wang YS, Huang NK, Lin YC, Chang WC, Huang WC. Aspirin and Sulindac act via different mechanisms to inhibit store-operated calcium channel: Implications for colorectal cancer metastasis. Biomed Pharmacother 2021; 145:112476. [PMID: 34864310 DOI: 10.1016/j.biopha.2021.112476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/08/2023] Open
Abstract
Store-operated Ca2+ channel (SOC)-regulated Ca2+ entry is involved in inflammation and colorectal cancer (CRC) progression, but clinically applicable treatments targeting this mechanism are lacking. Recent studies have shown that nonsteroidal anti-inflammatory drugs (NSAIDs) not only inhibit inflammation but they also suppress Ca2+ entry via SOC (SOCE). Therefore, delineating the mechanisms of SOCE inhibition by NSAIDs may lead to new CRC treatments. In this study, we tested eight candidate NSAIDs in Ca2+ imaging experiments and found that Aspirin and Sulindac were the most effective at suppressing SOCE. Furthermore, time-lapse FRET imaging using TIRF microscopy and ground state depletion (GSD) super-resolution (SR) imaging revealed that SOC was inhibited by Aspirin and Sulindac via different mechanisms. Aspirin quickly interrupted the STIM1-Orai1 interaction, whereas Sulindac mainly suppressed STIM1 translocation. Additionally, Aspirin and Sulindac both inhibited metastasis-related endpoints in CRC cells. Both drugs were used throughout the study at doses that suppressed CRC cell migration and invasion without altering cell survival. This is the first study to reveal the differential inhibitory mechanisms of Aspirin and Sulindac on SOC activity. Thus, our results shed new light on the therapeutic potential of Aspirin for CRC and SOCE-related diseases.
Collapse
Affiliation(s)
- Yu-Shiuan Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Xinyi District, Taipei 110, Taiwan, ROC
| | - Nai-Kuei Huang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Xinyi District, Taipei 110, Taiwan, ROC; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Beitou District, Taipei 112, Taiwan, ROC
| | - Yu-Chiao Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Zhongzheng District, Taipei 100, Taiwan, ROC
| | - Wei-Chiao Chang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Xinyi District, Taipei 110, Taiwan, ROC; Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Xinyi District, Taipei 110, Taiwan, ROC; Department of Pharmacy, Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Wenshan District, Taipei 116, Taiwan, ROC; Department of Pharmacology, National Defense Medical Center, Neihu District, Taipei 114, Taiwan, ROC.
| | - Wan-Chen Huang
- Single-Molecule Biology Core Lab, Institute of Cellular and Organismic Biology, Academia Sinica, Nankang District, Taipei 115, Taiwan, ROC; Institute of Medical Device and Imaging, National Taiwan University, Zhongzheng District, Taipei 100, Taiwan, ROC.
| |
Collapse
|
20
|
Zhang Q, He Y, Xu H, Li L, Guo Y, Zhang J, Cheng L, Yu H, Dai Y, Yang Q, Yang Z, Li C, Zhang S, Zhu S, Luo B, Gao Y. Modulation of STIM1 by a risk insertion/deletion polymorphism underlying genetics susceptibility to sudden cardiac death originated from coronary artery disease. Forensic Sci Int 2021; 328:111010. [PMID: 34592581 DOI: 10.1016/j.forsciint.2021.111010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022]
Abstract
Stromal interaction molecule 1 (STIM1), as a dynamic calcium signal transducer and key regulator of cardiomyocyte Ca2+ homeostasis, has been implicated in various pathological processes related to sudden cardiac death originated from coronary artery disease (SCD-CAD). In this study, we performed a systematic variant screening on promoter region of STIM1 to filter potential functional genetic variations. Based on the screening results, a 5-bp insertion/deletion (indel) polymorphism (rs3061890) in promoter region of STIM1 was selected as the candidate variant. We investigated the association of rs3061890 with SCD-CAD susceptibility in Chinese Han populations. The homozygote del/del genotype significantly increased risk for SCD-CAD as compared with the ins/ins genotype (odds ratio, 2.86 [95% confidence interval, 1.69-4.29]; P = 2.3 × 10-5). Compared with the common allele, the 5-bp deletion risk allele exhibited lower transcriptional capacity in luciferase assays. Intriguingly, genotype-phenotype correlation studies using human myocardium tissue samples revealed that the expression of STIM1 was associated with the genotype of rs3061890. Computational prediction combined with electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays provided convincing evidence for stronger binding affinity of ELF1 (E74 like ETS transcription factor 1) with the deletion allele promoter. Taken together, our findings implied an allele-specific mechanism of regulating the transcription of STIM1 via ELF1, which contribute to SCD-CAD susceptibility. rs3061890 may thus considered as a candidate genetic marker for SCD-CAD prediction and prevention.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Yan He
- Department of Epidemiology, Medical College of Soochow University, Suzhou, China.
| | - Hongfei Xu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Lijuan Li
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Yadong Guo
- Department of Forensic Sciences, School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Jianhua Zhang
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China.
| | - Lei Cheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Huan Yu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Yunda Dai
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Qi Yang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Zhenzhen Yang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Chengtao Li
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China.
| | - Suhua Zhang
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China.
| | - Shaohua Zhu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| | - Bin Luo
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Yuzhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
21
|
Jones CA, Hazlehurst LA. Role of Calcium Homeostasis in Modulating EMT in Cancer. Biomedicines 2021; 9:1200. [PMID: 34572386 PMCID: PMC8471317 DOI: 10.3390/biomedicines9091200] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Calcium is essential for cells to perform numerous physiological processes. In cancer, the augmentation of calcium signaling supports the more proliferative and migratory cells, which is a characteristic of the epithelial-to-mesenchymal transition (EMT). By genetically and epigenetically modifying genes, channels, and entire signaling pathways, cancer cells have adapted to survive with an extreme imbalance of calcium that allows them to grow and metastasize in an abnormal manner. This cellular remodeling also allows for the evasion of immune surveillance and the development of drug resistance, which lead to poor prognosis in patients. Understanding the role calcium flux plays in driving the phenotypes associated with invasion, immune suppression, metastasis, and drug resistance remains critical for determining treatments to optimize clinical outcomes and future drug discovery.
Collapse
Affiliation(s)
| | - Lori A. Hazlehurst
- Pharmaceutical and Pharmacological Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
22
|
Feng G, Wu J, Yang HL, Mu L. Discovery of Antioxidant Peptides from Amphibians: A Review. Protein Pept Lett 2021; 28:1220-1229. [PMID: 34493183 DOI: 10.2174/0929866528666210907145634] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/08/2021] [Accepted: 07/08/2021] [Indexed: 01/19/2023]
Abstract
In recent years, bioactive peptide drugs have attracted growing attention due to the increasing difficulty in developing new drugs with novel chemical structures. In addition, many diseases are linked to excessive oxidation in the human body. Therefore, the role of peptides with antioxidant activity in counteracting diseases related to oxidative stress is worth exploring. Amphibians are a major repository for bioactive peptides that protect the skin from biotic and abiotic stresses, such as microbial infection and radiation injury. We characterized the first amphibian- derived gene-encoded antioxidant peptides in 2008. Since then, a variety of antioxidant peptides have been detected in different amphibian species. In this work, the physicochemical properties of antioxidant peptides identified from amphibians are reviewed for the first time, particularly acquisition methods, amino acid characteristics, antioxidant mechanisms, and application prospects. This review should provide a reference for advancing the identification, structural analysis, and potential therapeutic value of natural antioxidant peptides.
Collapse
Affiliation(s)
- Guizhu Feng
- School of Basic Medical Sciences, Kunming Medical University, Kunming Yunnan 650500,China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming Yunnan 650500,China
| | - Hai-Long Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming Yunnan 650500,China
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, Kunming Yunnan 650500,China
| |
Collapse
|
23
|
Zhu J, Zhang X, Xie H, Wang Y, Zhang X, Lin Z. Cardiomyocyte Stim1 Deficiency Exacerbates Doxorubicin Cardiotoxicity by Magnification of Endoplasmic Reticulum Stress. J Inflamm Res 2021; 14:3945-3958. [PMID: 34421306 PMCID: PMC8373307 DOI: 10.2147/jir.s304520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/08/2021] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Doxorubicin (Dox) is an effective anticancer agent; however, its cardiotoxicity remains a challenge. Dysfunction of intracellular calcium ion (Ca2+) is implicated in the process of Dox-induced cardiomyocyte apoptosis. Although store-operated Ca2+ entry (SOCE) is suggested to be responsible for Ca2+ entry in cardiomyocytes, the direct role of store-operated Ca2+ channels in Dox-related cardiomyocyte apoptosis is unknown. MATERIALS AND METHODS Cardiomyocyte Stim1-specific knockout or overexpression mice were treated with Dox. Cardiomyocytes were pretreated with Stim1 adenovirus or siRNA followed by Dox incubation in vitro. Cardiac function and underlying mechanisms echocardiography were assessed via immunofluorescence, flow cytometry, real-time PCR, Western blotting and immunoprecipitation. RESULTS We observed the inhibition of Stim1 expression, association of Stim1 to Orai1 or Trpc1, and SOCE in Dox-treated mouse myocardium and cardiomyocytes. Orai1 and Trpc1 expression remained unchanged. Cardiomyocyte-specific deficiency of Stim1 exacerbated Dox-induced cardiac dysfunction and myocardial apoptosis. However, specific overexpression of Stim1 in the myocardium was associated with amelioration of cardiac dysfunction and myocardial apoptosis. In vitro, STIM1 knockdown potentiated Dox-induced AC16 human cardiomyocyte apoptosis. This apoptosis was attenuated by STIM1 upregulation. Moreover, STIM1 downregulation enhanced Dox-induced endoplasmic reticulum (ER) stress in cardiomyocytes. In contrast, STIM1 overexpression inhibited the activation of the above molecular markers of ER stress. Immunoprecipitation assay showed that STIM1 interacted with GRP78 in cardiomyocytes. This interaction was attenuated in response to Dox treatment. CONCLUSION Our data demonstrate that cardiomyocyte STIM1 binding to GRP78 ameliorates Dox cardiotoxicity by inhibiting pro-apoptotic ER stress.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215008, Jiangsu, People’s Republic of China
| | - Xia Zhang
- Department of Anesthesiology, Wuzhong People’s Hospital, Suzhou, Jiangsu, 215128, People’s Republic of China
| | - Hong Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215008, Jiangsu, People’s Republic of China
| | - Yuye Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215008, Jiangsu, People’s Republic of China
| | - Xiaoxiao Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215008, Jiangsu, People’s Republic of China
| | - Zhaoheng Lin
- Intensive Care Unit, People’s Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Jinghong City, 666100, Yunnan, People’s Republic of China
| |
Collapse
|
24
|
Luo Y, Li Z, Ge P, Guo H, Li L, Zhang G, Xu C, Chen H. Comprehensive Mechanism, Novel Markers and Multidisciplinary Treatment of Severe Acute Pancreatitis-Associated Cardiac Injury - A Narrative Review. J Inflamm Res 2021; 14:3145-3169. [PMID: 34285540 PMCID: PMC8286248 DOI: 10.2147/jir.s310990] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is one of the common acute abdominal inflammatory diseases in clinic with acute onset and rapid progress. About 20% of the patients will eventually develop into severe acute pancreatitis (SAP) characterized by a large number of inflammatory cells infiltration, gland flocculus flaky necrosis and hemorrhage, finally inducing systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS). Pancreatic enzyme activation, intestinal endotoxemia (IETM), cytokine activation, microcirculation disturbance, autonomic nerve dysfunction and autophagy dysregulation all play an essential role in the occurrence and progression of SAP. Organ dysfunction is the main cause of early death in SAP. Acute kidney injury (AKI) and acute lung injury (ALI) are common, while cardiac injury (CI) is not, but the case fatality risk is high. Many basic studies have observed obvious ultrastructure change of heart in SAP, including myocardial edema, cardiac hypertrophy, myocardial interstitial collagen deposition. Moreover, in clinical practice, patients with SAP often presented various abnormal electrocardiogram (ECG) and cardiac function. Cases complicated with acute myocardial infarction and pericardial tamponade have also been reported and even result in stress cardiomyopathy. Due to the molecular mechanisms underlying SAP-associated cardiac injury (SACI) remain poorly understood, and there is no complete, unified treatment and sovereign remedy at present, this article reviews reports referring to the pathogenesis, potential markers and treatment methods of SACI in recent years, in order to improve the understanding of cardiac injury in severe pancreatitis.
Collapse
Affiliation(s)
- YaLan Luo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - ZhaoXia Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Peng Ge
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaoYa Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - GuiXin Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - CaiMing Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaiLong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
25
|
Zheng CB, Gao WC, Xie M, Li Z, Ma X, Song W, Luo D, Huang Y, Yang J, Zhang P, Huang Y, Yang W, Yao X. Ang II Promotes Cardiac Autophagy and Hypertrophy via Orai1/STIM1. Front Pharmacol 2021; 12:622774. [PMID: 34079454 PMCID: PMC8165566 DOI: 10.3389/fphar.2021.622774] [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: 10/29/2020] [Accepted: 03/16/2021] [Indexed: 01/07/2023] Open
Abstract
The pathophysiology of cardiac hypertrophy is complex and multifactorial. Both the store-operated Ca2+ entry (SOCE) and excessive autophagy are the major causative factors for pathological cardiac hypertrophy. However, it is unclear whether these two causative factors are interdependent. In this study, we examined the functional role of SOCE and Orai1 in angiotensin II (Ang II)-induced autophagy and hypertrophy using in vitro neonatal rat cardiomyocytes (NRCMs) and in vivo mouse model, respectively. We show that YM-58483 or SKF-96365 mediated pharmacological inhibition of SOCE, or silencing of Orai1 with Orail-siRNA inhibited Ang II-induced cardiomyocyte autophagy both in vitro and in vivo. Also, the knockdown of Orai1 attenuated Ang II-induced pathological cardiac hypertrophy. Together, these data suggest that Ang II promotes excessive cardiomyocyte autophagy through SOCE/Orai1 which can be the prime contributing factors in cardiac hypertrophy.
Collapse
Affiliation(s)
- Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Wen-Cong Gao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Mingxu Xie
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Zhichao Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Xin Ma
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Wencong Song
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dan Luo
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Yongxiang Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Jichen Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Peng Zhang
- Longgang E.N.T. Hospital and Shenzhen Key Laboratory of E.N.T., Shenzhen, China
| | - Yu Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
26
|
Lele W, Lei L, Liting Q. Resveratrol sensitizes A549 cells to irradiation damage via suppression of store-operated calcium entry with Orai1 and STIM1 downregulation. Exp Ther Med 2021; 21:587. [PMID: 33850559 PMCID: PMC8027717 DOI: 10.3892/etm.2021.10019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 11/26/2020] [Indexed: 01/04/2023] Open
Abstract
Resveratrol is a natural polyphenol with multiple positive biofunctions and was found to have potential as a radiosensitizer with an intricate molecular mechanism. Store-operated calcium entry (SOCE) is a novel intracellular calcium regulatory pattern that is mainly mediated by iron channels, such as by the stromal interaction molecule (STIM) and calcium release-activated calcium channel protein (Orai) families. SOCE was recently reported to be suppressed via the downregulation of STIM or Orai families for the promotion of tumor cell death induced by resveratrol. In the present study, resveratrol combined with irradiation treatment were found to induce more evident cell damage compared with irradiation treatment alone, as shown with Cell Counting Kit-8 assay and mitochondrial membrane potential detection with rhodamine 123. Additionally, resveratrol combined with irradiation treatment decreased the expression of STIM1 and Orai1, while it had no effects on STIM2, Orai2 and Orai3. Moreover, resveratrol combined with irradiation treatment lead to alleviated thapsigargin-induced SOCE. In addition, overexpression of STIM1 and Orai1 reversed resveratrol-induced SOCE inhibition and reduced death in A549 cells under irradiation. In summary, the present results revealed that resveratrol can significantly enhance the effect of irradiation damage on lung adenocarcinoma A549 cells, and this effect may be mediated by suppression of SOCE with reduced expression of both STIM1 and Orai1.
Collapse
Affiliation(s)
- Wu Lele
- Department of General Medicine, First People's Hospital of Yuhang, Hangzhou, Zhejiang 311100, P.R. China.,Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| | - Lv Lei
- Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China.,Epigenetic Laboratory, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| | - Qian Liting
- Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China.,Epigenetic Laboratory, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
27
|
Cavus O, Williams J, Musa H, El Refaey M, Gratz D, Shaheen R, Schwieterman NA, Koenig S, Antwi-Boasiako S, Young LJ, Xu X, Han M, Wold LE, Hund TJ, Mohler PJ, Bradley EA. Giant ankyrin-G regulates cardiac function. J Biol Chem 2021; 296:100507. [PMID: 33675749 PMCID: PMC8040283 DOI: 10.1016/j.jbc.2021.100507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022] Open
Abstract
Cardiovascular disease (CVD) remains the most common cause of adult morbidity and mortality in developed nations. As a result, predisposition for CVD is increasingly important to understand. Ankyrins are intracellular proteins required for the maintenance of membrane domains. Canonical ankyrin-G (AnkG) has been shown to be vital for normal cardiac function, specifically cardiac excitability, via targeting and regulation of the cardiac voltage-gated sodium channel. Noncanonical (giant) AnkG isoforms play a key role in neuronal membrane biogenesis and excitability, with evidence for human neurologic disease when aberrant. However, the role of giant AnkG in cardiovascular tissue has yet to be explored. Here, we identify giant AnkG in the myocardium and identify that it is enriched in 1-week-old mice. Using a new mouse model lacking giant AnkG expression in myocytes, we identify that young mice displayed a dilated cardiomyopathy phenotype with aberrant electrical conduction and enhanced arrhythmogenicity. Structural and electrical dysfunction occurred at 1 week of age, when giant AnkG was highly expressed and did not appreciably change in adulthood until advanced age. At a cellular level, loss of giant AnkG results in delayed and early afterdepolarizations. However, surprisingly, giant AnkG cKO myocytes display normal INa, but abnormal myocyte contractility, suggesting unique roles of the large isoform in the heart. Finally, transcript analysis provided evidence for unique pathways that may contribute to the structural and electrical findings shown in giant AnkG cKO animals. In summary, we identify a critical role for giant AnkG that adds to the diversity of ankyrin function in the heart.
Collapse
Affiliation(s)
- Omer Cavus
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jordan Williams
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Hassan Musa
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Mona El Refaey
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Dan Gratz
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Rebecca Shaheen
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Neill A Schwieterman
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sara Koenig
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Steve Antwi-Boasiako
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Lindsay J Young
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Xianyao Xu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Mei Han
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Loren E Wold
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Elisa A Bradley
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
28
|
Rosenberg P, Zhang H, Bryson VG, Wang C. SOCE in the cardiomyocyte: the secret is in the chambers. Pflugers Arch 2021; 473:417-434. [PMID: 33638008 PMCID: PMC7910201 DOI: 10.1007/s00424-021-02540-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 11/24/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is an ancient and ubiquitous Ca2+ signaling pathway that is present in virtually every cell type. Over the last two decades, many studies have implicated this non-voltage dependent Ca2+ entry pathway in cardiac physiology. The relevance of the SOCE pathway in cardiomyocytes is often questioned given the well-established role for excitation contraction coupling. In this review, we consider the evidence that STIM1 and SOCE contribute to Ca2+ dynamics in cardiomyocytes. We discuss the relevance of this pathway to cardiac growth in response to developmental and pathologic cues. We also address whether STIM1 contributes to Ca2+ store refilling that likely impacts cardiac pacemaking and arrhythmogenesis in cardiomyocytes.
Collapse
Affiliation(s)
- Paul Rosenberg
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27705, USA.
| | - Hengtao Zhang
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27705, USA
| | | | - Chaojian Wang
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27705, USA
| |
Collapse
|
29
|
Liu X, Pan Z. Store-Operated Calcium Entry in the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:303-333. [DOI: 10.1007/978-981-16-4254-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Liu CC, Miao Y, Chen RL, Zhang YQ, Wu H, Yang SM, Shang LQ. STIM1 mediates IAV-induced inflammation of lung epithelial cells by regulating NLRP3 and inflammasome activation via targeting miR-223. Life Sci 2020; 266:118845. [PMID: 33278394 DOI: 10.1016/j.lfs.2020.118845] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/26/2022]
Abstract
AIMS Influenza A virus (IAV) infection accelerates the inflammatory injury of lung epithelial cells that contributes to pulmonary lesion. Recently, stromal interaction molecule 1 (STIM1) was found to mediate cellular immune response and participated in lung tumorigenesis. Our study aimed to illustrate the function and mechanism of STIM1 in IAV-induced inflammation injury and oxidative stress of lung epithelial cells. MAIN METHODS We evaluated the levels of STIM1 in IAV-infected patients' serum and BEAS-2B cells using RT-qPCR, Elisa and western blotting methods. MTT and Elisa were performed to measure cell viability and cytokine contents. Besides, ROS intensity, SOD contents and cell apoptosis were detected based on DCFH-DA probe, colorimetry and cell death kits. A luciferase assay and Pearson's correlation analysis evaluated the associations between target genes. KEY FINDINGS STIM1 was dramatically up-regulated in IAV-infected patients' serum and BEAS-2B cells. Silencing STIM1 in vitro inhibited oxidative stress and inflammatory responses induced by IAV, and reversed cell viability and suppressed apoptosis. Moreover, miR-223 and NLRP3 were negatively and positively correlated with STIM1. STIM1 was found to regulate NLRP3 expression by binding the AACUGAC motif in miR-223. STIM1/miR-223/NLRP3 axis modulated IAV-induced inflammation injury of lung epithelial cells. SIGNIFICANCE Our evidence indicated that silencing STIM1 alleviated IAV-induced inflammation injury of lung epithelial cells by inactivating NLRP3 and inflammasome via promoting miR-223 expression. These findings may contribute to understand the mechanism of IAV-induced lung injury and help for therapy of IAV infection.
Collapse
Affiliation(s)
- Cui-Cui Liu
- Department of Respiratory and Asthma, Xi'an Children's Hospital, Xi'an 710003, China
| | - Yi Miao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Rui-Lin Chen
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Yong-Qing Zhang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Hua Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Shu-Mei Yang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Li-Qun Shang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China.
| |
Collapse
|
31
|
Zhang I, Hu H. Store-Operated Calcium Channels in Physiological and Pathological States of the Nervous System. Front Cell Neurosci 2020; 14:600758. [PMID: 33328896 PMCID: PMC7732603 DOI: 10.3389/fncel.2020.600758] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Store-operated calcium channels (SOCs) are widely expressed in excitatory and non-excitatory cells where they mediate significant store-operated calcium entry (SOCE), an important pathway for calcium signaling throughout the body. While the activity of SOCs has been well studied in non-excitable cells, attention has turned to their role in neurons and glia in recent years. In particular, the role of SOCs in the nervous system has been extensively investigated, with links to their dysregulation found in a wide variety of neurological diseases from Alzheimer’s disease (AD) to pain. In this review, we provide an overview of their molecular components, expression, and physiological role in the nervous system and describe how the dysregulation of those roles could potentially lead to various neurological disorders. Although further studies are still needed to understand how SOCs are activated under physiological conditions and how they are linked to pathological states, growing evidence indicates that SOCs are important players in neurological disorders and could be potential new targets for therapies. While the role of SOCE in the nervous system continues to be multifaceted and controversial, the study of SOCs provides a potentially fruitful avenue into better understanding the nervous system and its pathologies.
Collapse
Affiliation(s)
- Isis Zhang
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Huijuan Hu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
32
|
Li Y, Yi J, Liu W, Liu Y, Liu J. Gaining insight into cellular cardiac physiology using single particle tracking. J Mol Cell Cardiol 2020; 148:63-77. [PMID: 32871158 DOI: 10.1016/j.yjmcc.2020.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 11/29/2022]
Abstract
Single particle tracking (SPT) is a robust technique to monitor single-molecule behaviors in living cells directly. By this approach, we can uncover the potential biological significance of particle dynamics by statistically characterizing individual molecular behaviors. SPT provides valuable information at the single-molecule level, that could be obscured by simple averaging that is inherent to conventional ensemble measurements. Here, we give a brief introduction to SPT including the commonly used optical implementations, fluorescence labeling strategies, and data analysis methods. We then focus on how SPT has been harnessed to decipher myocardial function. In this context, SPT has provided novel insight into the lateral diffusion of signal receptors and ion channels, the dynamic organization of cardiac nanodomains, subunit composition and stoichiometry of cardiac ion channels, myosin movement along actin filaments, the kinetic features of transcription factors involved in cardiac remodeling, and the intercellular communication by nanotubes. Finally, we speculate on the prospects and challenges of applying SPT to future questions regarding cellular cardiac physiology using SPT.
Collapse
Affiliation(s)
- Ying Li
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| | - Jing Yi
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| | - Wenjuan Liu
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| | - Yun Liu
- The Seventh Affiliated Hospital, Sun Yat-sen University, Guangdong Province, China.
| | - Jie Liu
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
33
|
Gavali JT, Carrillo ED, García MC, Sánchez JA. The mitochondrial K-ATP channel opener diazoxide upregulates STIM1 and Orai1 via ROS and the MAPK pathway in adult rat cardiomyocytes. Cell Biosci 2020; 10:96. [PMID: 32817784 PMCID: PMC7424994 DOI: 10.1186/s13578-020-00460-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/07/2020] [Indexed: 11/15/2022] Open
Abstract
Background Openers of mitochondrial adenosine triphosphate-dependent potassium (mKATP) channels like diazoxide increase reactive oxygen species (ROS) production in cardiac cells and reduce Ca2+ elevations produced by ischemia–reperfusion, protecting the heart from damage. In this study we tested the hypothesis that opening mKATP channels regulates expression of the major components of store-operated Ca2+ entry (SOCE) STIM1 and Orai1. Results Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot experiments showed that diazoxide increased expression of STIM1 and Orai1 at the mRNA and protein levels, respectively, in adult rat cardiomyocytes. Immunofluorescence analyses revealed that diazoxide also disrupted the striated distribution pattern of STIM1. These effects were prevented by the ROS scavenger N-acetyl cysteine (NAC), the mKATP channel antagonist 5-hydroxydecanoate (5-HD), or the protein synthesis inhibitor cycloheximide (CHX). Confocal microscopy revealed that diazoxide also led to nuclear translocation of the transcription factors c-Fos and NFκB, which was also blocked by NAC or 5-HD. Finally, the MAPK pathway inhibitor UO126 attenuated diazoxide-induced upregulation of STIM1 and Orai1 expression. Conclusions Our results suggest that opening mitochondrial potassium ATP channels with diazoxide upregulates the expression of STIM1 and Orai1 by de novo synthesis by a mechanism that involves NFkB, c-Fos, and ROS via MAPK/ERK signaling.
Collapse
Affiliation(s)
- Joice T Gavali
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, 07360 Ciudad de México, CDMX Mexico
| | - Elba D Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, 07360 Ciudad de México, CDMX Mexico
| | - María C García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, 07360 Ciudad de México, CDMX Mexico
| | - Jorge A Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, 07360 Ciudad de México, CDMX Mexico
| |
Collapse
|
34
|
Segin S, Berlin M, Richter C, Medert R, Flockerzi V, Worley P, Freichel M, Camacho Londoño JE. Cardiomyocyte-Specific Deletion of Orai1 Reveals Its Protective Role in Angiotensin-II-Induced Pathological Cardiac Remodeling. Cells 2020; 9:cells9051092. [PMID: 32354146 PMCID: PMC7290784 DOI: 10.3390/cells9051092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Pathological cardiac remodeling correlates with chronic neurohumoral stimulation and abnormal Ca2+ signaling in cardiomyocytes. Store-operated calcium entry (SOCE) has been described in adult and neonatal murine cardiomyocytes, and Orai1 proteins act as crucial ion-conducting constituents of this calcium entry pathway that can be engaged not only by passive Ca2+ store depletion but also by neurohumoral stimuli such as angiotensin-II. In this study, we, therefore, analyzed the consequences of Orai1 deletion for cardiomyocyte hypertrophy in neonatal and adult cardiomyocytes as well as for other features of pathological cardiac remodeling including cardiac contractile function in vivo. Cellular hypertrophy induced by angiotensin-II in embryonic cardiomyocytes from Orai1-deficient mice was blunted in comparison to cells from litter-matched control mice. Due to lethality of mice with ubiquitous Orai1 deficiency and to selectively analyze the role of Orai1 in adult cardiomyocytes, we generated a cardiomyocyte-specific and temporally inducible Orai1 knockout mouse line (Orai1CM–KO). Analysis of cardiac contractility by pressure-volume loops under basal conditions and of cardiac histology did not reveal differences between Orai1CM–KO mice and controls. Moreover, deletion of Orai1 in cardiomyocytes in adult mice did not protect them from angiotensin-II-induced cardiac remodeling, but cardiomyocyte cross-sectional area and cardiac fibrosis were enhanced. These alterations in the absence of Orai1 go along with blunted angiotensin-II-induced upregulation of the expression of Myoz2 and a lack of rise in angiotensin-II-induced STIM1 and Orai3 expression. In contrast to embryonic cardiomyocytes, where Orai1 contributes to the development of cellular hypertrophy, the results obtained from deletion of Orai1 in the adult myocardium reveal a protective function of Orai1 against the development of angiotensin-II-induced cardiac remodeling, possibly involving signaling via Orai3/STIM1-calcineurin-NFAT related pathways.
Collapse
Affiliation(s)
- Sebastian Segin
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Michael Berlin
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Christin Richter
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
| | - Rebekka Medert
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany;
| | - Paul Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA;
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Juan E. Camacho Londoño
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; (S.S.); (M.B.); (C.R.); (R.M.); (M.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-54-86863; Fax: +49-6221-54-8644
| |
Collapse
|
35
|
Li L, Li YQ, Sun ZW, Xu CM, Wu J, Liu GL, Bakheet AMH, Chen HL. Qingyi decoction protects against myocardial injuries induced by severe acute pancreatitis. World J Gastroenterol 2020; 26:1317-1328. [PMID: 32256019 PMCID: PMC7109276 DOI: 10.3748/wjg.v26.i12.1317] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/21/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We studied the protective effects of Qingyi decoction (QYD) (a Traditional Chinese Medicine) against severe acute pancreatitis (SAP)-induced myocardial infarction (MI).
AIM To study the function and mechanism of QYD in the treatment of myocardial injuries induced by SAP.
METHODS Ultrasonic cardiography, hematoxylin and eosin staining, immunohistochemistry, qRT-PCR, western blot, enzyme-linked immunosorbent assays, and apoptosis staining techniques were used to determine the effects of QYD following SAP-induced MI in Sprague-Dawley rats.
RESULTS Our SAP model showed severe myocardial histological abnormalities and marked differences in the symptoms, mortality rate, and ultrasonic cardiography outputs among the different groups compared to the control. The expression of serum cytokines [interleukin (IL)-1ß, IL-6, IL-8, IL-12, amyloid β, and tumor necrosis factor-α] were significantly higher in the SAP versus QYD treated group (P < 0.05 for all). STIM1 and Orai1 expression in myocardial tissue extracts were significantly decreased post QYD gavage (P < 0.001). There was no significant histological difference between the 2-aminoethyl diphenylborinate inhibitor and QYD groups. The SAP group had a significantly higher apoptosis index score compared to the QYD group (P < 0.001).
CONCLUSION QYD conferred cardio-protection against SAP-induced MI by regulating myocardial-associated protein expression (STIM1 and Orai1).
Collapse
Affiliation(s)
- Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, Liaoning Province, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| | - Yong-Qi Li
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 3058575, Japan
| | - Zhong-Wei Sun
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
- Department of Emergency Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Cai-Ming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
- Department of Traditional Chinese Medicine, Dalian Obstetrics and Gynecology Hospital, Dalian 116021, Liaoning Province, China
| | - Jun Wu
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, Liaoning Province, China
| | - Ge-Liang Liu
- Department of Urology Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, Liaoning Province, China
| | - Ahmed MH Bakheet
- Department of Pathology, The Third Affiliated Hospital of San Yet-sen University, Guangzhou 510360, Guangdong Province, China
| | - Hai-Long Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| |
Collapse
|
36
|
Demirci T, Bilge N, Ucar M, Abuc OO, Atilay H. Electron Microscopic and Immunohistochemical Examination of the Effect of 2-Aminoethoxydiphenyl Borate on Optic Nerve Injury in A Rat Model. Eurasian J Med 2020; 52:61-66. [PMID: 32158317 DOI: 10.5152/eurasianjmed.2020.19089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective We conducted this study to explore the possible protective effect of 2-aminoethoxydiphenyl borate (2-APB) on experimentally induced optic nerve injury in an acute ischemia-reperfusion (AIR) model. Materials and Methods A total of 30 Wistar albino rats were randomly divided into sham, AIR, and AIR+treatment (AIR10) groups. In the sham group, AIR model was not created. In the AIR group, AIR model was created without the administration of drug. In the AIR10 group, 2-APB was administered 10 min before reperfusion. Results Tissue samples were subjected to histological, immunohistochemical, and electron microscopic procedures. Histopathological examination revealed intense hypertrophic cells, more glial cells, capillary dilatation, and intense demyelination areas in the AIR group compared to those in the sham and AIR10 groups. Immunohistochemical staining demonstrated an increase in Orai1 and STIM1 immunoreactivity in the AIR group but less intense staining in the AIR10 group. Electron microscopy revealed injury in optic nerve axons in the AIR group, whereas this type of injury occurred to a lesser extent in the AIR10 group. Conclusion In rats, store-operated Ca2+ entry in the cell had an essential role in optic nerve ischemia-reperfusion injury, and 2-ABP may have a protective effect on optic nerve injury caused due to AIR.
Collapse
Affiliation(s)
- Tuba Demirci
- Department of Histology and Embryology, Atatürk University School of Medicine, Erzurum, Turkey
| | - Nuray Bilge
- Department of Neurology, Ataturk University School of Medicine, Erzurum, Turkey
| | - Metin Ucar
- Department of Ophthalmology, Regional Training and Research Hospital, Erzurum, Turkey
| | - Ozlem Ozgul Abuc
- Department of Histology and Embryology, Atatürk University School of Medicine, Erzurum, Turkey
| | - Hilal Atilay
- Department of Histology and Embryology, Atatürk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
37
|
Zhang H, Bryson V, Luo N, Sun AY, Rosenberg P. STIM1-Ca 2+ signaling in coronary sinus cardiomyocytes contributes to interatrial conduction. Cell Calcium 2020; 87:102163. [PMID: 32014794 DOI: 10.1016/j.ceca.2020.102163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/11/2019] [Accepted: 01/17/2020] [Indexed: 11/19/2022]
Abstract
Pacemaker action potentials emerge from the sinoatrial node (SAN) and rapidly propagate through the atria to the AV node via preferential conduction pathways, including one associated with the coronary sinus. However, few distinguishing features of these tracts are known. Identifying specific molecular markers to distinguish among these conduction pathways will have important implications for understanding atrial conduction and atrial arrhythmogenesis. Using a Stim1 reporter mouse, we discovered stromal interaction molecule 1 (STIM1)-expressing coronary sinus cardiomyocytes (CSC)s in a tract from the SAN to the coronary sinus. Our studies here establish that STIM1 is a molecular marker of CSCs and we propose a role for STIM1-CSCs in interatrial conduction. Deletion of Stim1 from the CSCs slowed interatrial conduction and increased susceptibility to atrial arrhythmias. Store-operated Ca2+ currents (Isoc) in response to Ca2+ store depletion were markedly reduced in CSCs and their action potentials showed electrical remodeling. Our studies identify STIM1 as a molecular marker for a coronary sinus interatrial conduction pathway. We propose a role for SOCE in Ca2+ signaling of CSCs and implicate STIM1 in atrial arrhythmogenesis.
Collapse
Affiliation(s)
- Hengtao Zhang
- Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Box 103031 Med Ctr, Durham, NC, 27710, United States
| | - Victoria Bryson
- Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Box 103031 Med Ctr, Durham, NC, 27710, United States
| | - Nancy Luo
- Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Box 103031 Med Ctr, Durham, NC, 27710, United States
| | - Albert Y Sun
- Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Box 103031 Med Ctr, Durham, NC, 27710, United States
| | - Paul Rosenberg
- Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Box 103031 Med Ctr, Durham, NC, 27710, United States.
| |
Collapse
|
38
|
Sampieri R, Fuentes E, Carrillo ED, Hernández A, García MC, Sánchez JA. Pharmacological Preconditioning Using Diazoxide Regulates Store-Operated Ca 2 + Channels in Adult Rat Cardiomyocytes. Front Physiol 2020; 10:1589. [PMID: 32009985 PMCID: PMC6972595 DOI: 10.3389/fphys.2019.01589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/19/2019] [Indexed: 01/31/2023] Open
Abstract
Voltage-dependent Ca2+ channels and store-operated Ca2+ channels (SOCs) are the major routes of Ca2+ entry into mammalian cells. Previously, we reported that pharmacological preconditioning (PPC) leads to a decrease in the amplitude of L-type calcium channel current in the heart. In this study, we examined PPC-associated changes in SOC function. We measured adult cardiomyocyte membrane currents using the whole-cell patch-clamp technique, and we evaluated reactive oxygen species (ROS) production and intracellular Ca2+ levels in cardiomyocytes using fluorescent probes. Diazoxide (Dzx) and thapsigargin (Tg) were used to induce PPC and to deplete internal stores of Ca2+, respectively. Ca2+ store depletion generated inward currents with strong rectification, which were suppressed by the SOC blocker GSK-7975-A. These currents were completely abolished by PPC, an effect that could be countered with 5-hydroxydecanoate (5-HD; a selective mitochondrial ATP-sensitive K+ channel blocker), an intracellular mitochondrial energizing solution, or Ni2+ [a blocker of sodium-calcium exchanger (NCX)]. Buffering of ROS and intracellular Ca2+ also prevented PPC effects on SOC currents. Refilling of intracellular stores was largely suppressed by PPC, as determined by measuring intracellular Ca2+ with a fluorescent Ca2+ indicator. These results indicate that influx of Ca2+ through SOCs is inhibited by their ROS and Ca2+-dependent inactivation during PPC and that NCX is a likely source of PPC-inactivating Ca2+. We further showed that NCX associates with Orai1. Down-regulation of SOCs by PPC may play a role in cardioprotection following ischemia-reperfusion.
Collapse
Affiliation(s)
- Raúl Sampieri
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Eridani Fuentes
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Elba D Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Ascención Hernández
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - María C García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Jorge A Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| |
Collapse
|
39
|
Avila-Medina J, Mayoral-González I, Galeano-Otero I, Redondo PC, Rosado JA, Smani T. Pathophysiological Significance of Store-Operated Calcium Entry in Cardiovascular and Skeletal Muscle Disorders and Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:489-504. [PMID: 31646522 DOI: 10.1007/978-3-030-12457-1_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Store-Operated Ca2+ Entry (SOCE) is an important Ca2+ influx pathway expressed by several excitable and non-excitable cell types. SOCE is recognized as relevant signaling pathway not only for physiological process, but also for its involvement in different pathologies. In fact, independent studies demonstrated the implication of essential protein regulating SOCE, such as STIM, Orai and TRPCs, in different pathogenesis and cell disorders, including cardiovascular disease, muscular dystrophies and angiogenesis. Compelling evidence showed that dysregulation in the function and/or expression of isoforms of STIM, Orai or TRPC play pivotal roles in cardiac hypertrophy and heart failure, vascular remodeling and hypertension, skeletal myopathies, and angiogenesis. In this chapter, we summarized the current knowledge concerning the mechanisms underlying abnormal SOCE and its involvement in some diseases, as well as, we discussed the significance of STIM, Orai and TRPC isoforms as possible therapeutic targets for the treatment of angiogenesis, cardiovascular and skeletal muscle diseases.
Collapse
Affiliation(s)
- Javier Avila-Medina
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Isabel Mayoral-González
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
- Department of Surgery, University of Seville, Sevilla, Spain
| | - Isabel Galeano-Otero
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Pedro C Redondo
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain.
- CIBERCV, Madrid, Spain.
| |
Collapse
|
40
|
Specific Upregulation of TRPC1 and TRPC5 Channels by Mineralocorticoid Pathway in Adult Rat Ventricular Cardiomyocytes. Cells 2019; 9:cells9010047. [PMID: 31878108 PMCID: PMC7017140 DOI: 10.3390/cells9010047] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/18/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023] Open
Abstract
Whereas cardiac TRPC (transient receptor potential canonical) channels and the associated store-operated Ca2+ entry (SOCE) are abnormally elevated during cardiac hypertrophy and heart failure, the mechanism of this upregulation is not fully elucidated but might be related to the activation of the mineralocorticoid pathway. Using a combination of biochemical, Ca2+ imaging, and electrophysiological techniques, we determined the effect of 24-h aldosterone treatment on the TRPCs/Orai-dependent SOCE in adult rat ventricular cardiomyocytes (ARVMs). The 24-h aldosterone treatment (from 100 nM to 1 µM) enhanced depletion-induced Ca2+ entry in ARVMs, as assessed by a faster reduction of Fura-2 fluorescence decay upon the addition of Mn2+ and increased Fluo-4/AM fluorescence following Ca2+ store depletion. These effects were prevented by co-treatment with a specific mineralocorticoid receptor (MR) antagonist, RU-28318, and they are associated with the enhanced depletion-induced N-[4-[3,5-Bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-4-methyl-1,2,3-thiadiazole-5-carboxamide (BTP2)-sensitive macroscopic current recorded by patch-clamp experiments. Molecular screening by qRT-PCR and Western blot showed a specific upregulation of TRPC1, TRPC5, and STIM1 expression at the messenger RNA (mRNA) and protein levels upon 24-h aldosterone treatment of ARVMs, corroborated by immunostaining. Our study provides evidence that the mineralocorticoid pathway specifically promotes TRPC1/TRPC5-mediated SOCE in adult rat cardiomyocytes.
Collapse
|
41
|
Simonowski A, Wilhelm T, Habib P, Zorn CN, Huber M. Differential use of BTK and PLC in FcεRI- and KIT-mediated mast cell activation: A marginal role of BTK upon KIT activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118622. [PMID: 31837347 DOI: 10.1016/j.bbamcr.2019.118622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/16/2019] [Accepted: 12/08/2019] [Indexed: 02/04/2023]
Abstract
In mast cells (MCs), the TEC family kinase (TFK) BTK constitutes a central regulator of antigen (Ag)-triggered, FcεRI-mediated PLCγ phosphorylation, Ca2+ mobilization, degranulation, and pro-inflammatory cytokine production. Less is known about the function of BTK in the context of stem cell factor (SCF)-induced KIT signaling. In bone marrow-derived MCs (BMMCs), Ag stimulation caused intense phosphorylation of BTK at Y551 in its active center and at Y223 in its SH3-domain, whereas in response to SCF only Y223 was significantly phosphorylated. Further data using the TFK inhibitor Ibrutinib indicated that BTK Y223 is phosphorylated by a non-BTK TFK upon SCF stimulation. In line, SCF-induced PLCγ1 phosphorylation was stronger attenuated by Ibrutinib than by BTK deficiency. Subsequent pharmacological analysis of PLCγ function revealed a total block of SCF-induced Ca2+ mobilization by PLC inhibition, whereas only the sustained phase of Ca2+ flux was curtailed in Ag-stimulated BMMCs. Despite this severe stimulus-dependent difference in inducing Ca2+ mobilization, PLCγ inhibition suppressed Ag- and SCF-induced degranulation and pro-inflammatory cytokine production to comparable extents, suggesting involvement of additional TFK(s) or PLCγ-dependent signaling components. In addition to PLCγ, the MAPKs p38 and JNK were activated by Ag in a BTK-dependent manner; this was not observed upon SCF stimulation. Hence, FcεRI and KIT employ different mechanisms for activating PLCγ, p38, and JNK, which might strengthen their cooperation regarding pro-inflammatory MC effector functions. Importantly, our data clearly demonstrate that analyzing BTK Y223 phosphorylation is not sufficient to prove BTK activation.
Collapse
Affiliation(s)
- Anne Simonowski
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Carolin N Zorn
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.
| |
Collapse
|
42
|
Xu H, Cheng J, Wang X, Liu H, Wang S, Wu J, Xu B, Chen A, He F. Resveratrol pretreatment alleviates myocardial ischemia/reperfusion injury by inhibiting STIM1-mediated intracellular calcium accumulation. J Physiol Biochem 2019; 75:607-618. [DOI: 10.1007/s13105-019-00704-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022]
|
43
|
Cacheux M, Strauss B, Raad N, Ilkan Z, Hu J, Benard L, Feske S, Hulot JS, Akar FG. Cardiomyocyte-Specific STIM1 (Stromal Interaction Molecule 1) Depletion in the Adult Heart Promotes the Development of Arrhythmogenic Discordant Alternans. Circ Arrhythm Electrophysiol 2019; 12:e007382. [PMID: 31726860 PMCID: PMC6867678 DOI: 10.1161/circep.119.007382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND STIM1 (stromal interaction molecule 1) is a calcium (Ca2+) sensor that regulates cardiac hypertrophy by triggering store-operated Ca2+ entry. Because STIM1 binding to phospholamban increases sarcoplasmic reticulum Ca2+ load independent of store-operated Ca2+ entry, we hypothesized that it controls electrophysiological function and arrhythmias in the adult heart. METHODS Inducible myocyte-restricted STIM1-KD (STIM1 knockdown) was achieved in adult mice using an αMHC (α-myosin heavy chain)-MerCreMer system. Mechanical and electrophysiological properties were examined using echocardiography in vivo and optical action potential (AP) mapping ex vivo in tamoxifen-induced STIM1flox/flox-Cretg/- (STIM1-KD) and littermate controls for STIM1flox/flox (referred to as STIM1-Ctl) and for Cretg/- without STIM deletion (referred to as Cre-Ctl). RESULTS STIM1-KD mice (N=23) exhibited poor survival compared with STIM1-Ctl (N=22) and Cre-Ctl (N=11) with >50% mortality after only 8-days of cardiomyocyte-restricted STIM1-KD. STIM1-KD but not STIM1-Ctl or Cre-Ctl hearts exhibited a proclivity for arrhythmic behavior, ranging from frequent ectopy to pacing-induced ventricular tachycardia/ventricular fibrillation (VT/VF). Examination of the electrophysiological substrate revealed decreased conduction velocity and increased AP duration (APD) heterogeneity in STIM1-KD. These features, however, were comparable in VT/VF(+) and VT/VF(-) hearts. We also uncovered a marked increase in the magnitude of APD alternans during rapid pacing, and the emergence of a spatially discordant alternans profile in STIM1-KD hearts. Unlike conduction velocity slowing and APD heterogeneity, the magnitude of APD alternans was greater (by 80%, P<0.05) in VT/VF(+) versus VT/VF(-) STIM1-KD hearts. Detailed phase mapping during the initial beats of VT/VF identified one or more rotors that were localized along the nodal line separating out-of-phase alternans regions. CONCLUSIONS In an adult murine model with inducible and myocyte-specific STIM1 depletion, we demonstrate for the first time the regulation of spatially discordant alternans by STIM1. Early mortality in STIM1-KD mice is likely related to enhanced susceptibility to VT/VF secondary to discordant APD alternans.
Collapse
Affiliation(s)
- Marine Cacheux
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Benjamin Strauss
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Nour Raad
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Zeki Ilkan
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Jun Hu
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Ludovic Benard
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine (S.F.)
| | - Jean-Sebastien Hulot
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Fadi G Akar
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| |
Collapse
|
44
|
Smani T, Gallardo-Castillo I, Ávila-Médina J, Jimenez-Navarro MF, Ordoñez A, Hmadcha A. Impact of Diabetes on Cardiac and Vascular Disease: Role of Calcium Signaling. Curr Med Chem 2019; 26:4166-4177. [DOI: 10.2174/0929867324666170523140925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 02/14/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Abstract
The pathophysiology linking diabetes and cardiovascular disease (CVD) is
complex and multifactorial. The specific type of cardiomyopathy associated with diabetes,
known as diabetic cardiomyopathy (DCM), is recognized as asymptomatic progression
of structural and functional remodeling in the heart of diabetic patients in the absence
of coronary atherosclerosis and hypertension. In other words, the presence of heart disease
specifically in diabetic patients is also known as diabetic heart disease. This article
reviews the impact of diabetes in heart and vascular beds focusing on molecular mechanisms
involving the oxidative stress, the inflammation, the endothelium dysfunction and
the alteration of the homeostasis of calcium, among others mechanisms. Understanding
these mechanisms will help identify and treat CVD in patients with diabetes, as well as to
plan efficient strategies to mitigate DCM impact in those patients.
Collapse
Affiliation(s)
- Tarik Smani
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | | | - Javier Ávila-Médina
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | - Manuel F. Jimenez-Navarro
- UGC del Corazon, Instituto de Biomedicina de Malaga (IBIMA), Hospital Clínico Universitario Virgen de la Victoria, Universidad de Malaga, Malaga, Spain
| | - Antonio Ordoñez
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Unversity of Pablo de Olavide- University of Seville-CSIC, Seville, Spain
| |
Collapse
|
45
|
Glucocorticoid stimulation increases cardiac contractility by SGK1-dependent SOCE-activation in rat cardiac myocytes. PLoS One 2019; 14:e0222341. [PMID: 31498847 PMCID: PMC6733454 DOI: 10.1371/journal.pone.0222341] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/27/2019] [Indexed: 01/28/2023] Open
Abstract
Aims Glucocorticoid (GC) stimulation has been shown to increase cardiac contractility by elevated intracellular [Ca] but the sources for Ca entry are unclear. This study aims to determine the role of store-operated Ca entry (SOCE) for GC-mediated inotropy. Methods and results Dexamethasone (Dex) pretreatment significantly increased cardiac contractile force ex vivo in Langendorff-perfused Sprague-Dawley rat hearts (2 mg/kg BW i.p. Dex 24 h prior to experiment). Moreover, Ca transient amplitude as well as fractional shortening were significantly enhanced in Fura-2-loaded isolated rat ventricular myocytes exposed to Dex (1 mg/mL Dex, 24 h). Interestingly, these Dex-dependent effects could be abolished in the presence of SOCE-inhibitors SKF-96356 (SKF, 2 μM) and BTP2 (5 μM). Ca transient kinetics (time to peak, decay time) were not affected by SOCE stimulation. Direct SOCE measurements revealed a negligible magnitude in untreated myocytes but a dramatic increase in SOCE upon Dex-pretreatment. Importantly, the Dex-dependent stimulation of SOCE could be blocked by inhibition of serum and glucocorticoid-regulated kinase 1 (SGK1) using EMD638683 (EMD, 50 μM). Dex preincubation also resulted in increased mRNA expression of proteins involved in SOCE (stromal interaction molecule 2, STIM2, and transient receptor potential cation channels 3/6, TRPC 3/6), which were also prevented in the presence of EMD. Conclusion Short-term GC-stimulation with Dex improves cardiac contractility by a SOCE-dependent mechanism, which appears to involve increased SGK1-dependent expression of the SOCE-related proteins. Since Ca transient kinetics were unaffected, SOCE appears to influence Ca cycling more by an integrated response across multiple cardiac cycles but not on a beat-to-beat basis.
Collapse
|
46
|
Shiou YL, Lin HT, Ke LY, Wu BN, Shin SJ, Chen CH, Tsai WC, Chu CS, Lee HC. Very Low-Density Lipoproteins of Metabolic Syndrome Modulates STIM1, Suppresses Store-Operated Calcium Entry, and Deranges Myofilament Proteins in Atrial Myocytes. J Clin Med 2019; 8:jcm8060881. [PMID: 31226824 PMCID: PMC6617489 DOI: 10.3390/jcm8060881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 01/16/2023] Open
Abstract
Individuals with metabolic syndrome (MetS) are at high risk for atrial myopathy and atrial fibrillation. Very low-density lipoproteins (VLDLs) of MetS (MetS-VLDLs) are cytotoxic to atrial myocytes in vivo and in vitro. The calcineurin-nuclear factor of activated T-cells (NFAT) pathway, which is regulated by stromal interaction molecule 1 (STIM1)/ calcium release-activated calcium channel protein 1 (Orai1)-mediated store-operated Ca2+ entry (SOCE), is a pivotal mediator of adaptive cardiac hypertrophy. We hypothesized that MetS-VLDLs could affect SOCE and the calcineurin-NFAT pathway. Normal-VLDL and MetS-VLDL samples were isolated from the peripheral blood of healthy volunteers and individuals with MetS. VLDLs were applied to HL-1 atrial myocytes for 18 h and were also injected into wild-type C57BL/6 male mouse tails three times per week for six weeks. After the sarcoplasmic reticulum (SR) Ca2+ store was depleted, SOCE was triggered upon reperfusion with 1.8 mM of Ca2+. SOCE was attenuated by MetS-VLDLs, along with reduced transcriptional and membranous expression of STIM1 (P = 0.025), and enhanced modification of O-GlcNAcylation on STIM1 protein, while Orai1 was unaltered. The nuclear translocation and activity of calcineurin were both reduced (P < 0.05), along with the alteration of myofilament proteins in atrial tissues. These changes were absent in normal-VLDL-treated cells. Our results demonstrated that MetS-VLDLs suppressed SOCE by modulating STIM1 at the transcriptional, translational, and post-translational levels, resulting in the inhibition of the calcineurin-NFAT pathway, which resulted in the alteration of myofilament protein expression and sarcomere derangement in atrial tissues. These findings may help explain atrial myopathy in MetS. We suggest a therapeutic target on VLDLs to prevent atrial fibrillation, especially for individuals with MetS.
Collapse
Affiliation(s)
- Yi-Lin Shiou
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsin-Ting Lin
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Liang-Yin Ke
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Bin-Nan Wu
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shyi-Jang Shin
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsiang-Chun Lee
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute/Center of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 807, Taiwan.
| |
Collapse
|
47
|
Malette J, Degrandmaison J, Giguère H, Berthiaume J, Frappier M, Parent JL, Auger-Messier M, Boulay G. MURC/CAVIN-4 facilitates store-operated calcium entry in neonatal cardiomyocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1249-1259. [PMID: 30951783 DOI: 10.1016/j.bbamcr.2019.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 01/29/2023]
Abstract
Intact store-operated calcium entry (SOCE) mechanisms ensure the maintenance of Ca2+ homeostasis in cardiomyocytes while their dysregulation promotes the development of cardiomyopathies. To better understand this calcium handling process in cardiomyocytes, we sought to identify unknown protein partners of stromal interaction molecule 1 (STIM1), a main regulatory protein of SOCE. We identified the muscle-related coiled-coil protein (MURC), also known as Cavin-4, as a candidate and showed that MURC interacts with STIM1 in cardiomyocytes. This interaction occurs via the HR1 and ERM domains of MURC and STIM1, respectively. Our results also demonstrated that the overexpression of MURC in neonatal rat ventricular myocytes (NRVM) is sufficient to potentiate SOCE and that its HR1 domain is required to mediate this effect. Interestingly, the R140W-MURC mutant, a missense variant of the HR1 domain associated with human dilated cardiomyopathy, exacerbates the SOCE increase in NRVM. Although the endogenous expression of STIM1 and Ca2+ channel Orai1 is not modulated under these conditions, we showed that MURC increases the interaction between these proteins under resting conditions. Our study provides novel evidence that MURC regulates SOCE by interacting with STIM1 in cardiomyocytes. In addition, we identified a first potential mechanism by which the R140W mutation of MURC may contribute to calcium mishandling and the development of cardiomyopathies.
Collapse
Affiliation(s)
- Julien Malette
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, QC J1H 5N4, Canada; Département de Médecine - Service de Cardiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jade Degrandmaison
- Département de Médecine - Service de Rhumatologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Hugo Giguère
- Département de Médecine - Service de Cardiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jonathan Berthiaume
- Département de Médecine - Service de Cardiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Maude Frappier
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, QC J1H 5N4, Canada
| | - Jean-Luc Parent
- Département de Médecine - Service de Rhumatologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Mannix Auger-Messier
- Département de Médecine - Service de Cardiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Guylain Boulay
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, QC J1H 5N4, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
48
|
Transient Receptor Potential Canonical Channel Blockers Improve Ventricular Contractile Functions After Ischemia/Reperfusion in a Langendorff-perfused Mouse Heart Model. J Cardiovasc Pharmacol 2019; 71:248-255. [PMID: 29389740 DOI: 10.1097/fjc.0000000000000566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reperfusion of ischemic myocardium is accompanied by intracellular Ca overload, leading to cardiac dysfunction. However, the mechanisms underlying intracellular Ca overload have yet to be fully elucidated. The mechanism may involve the activation of store-operated Ca entry, which is primarily mediated through the transient receptor potential canonical (TRPC) channels. This study was undertaken to examine the possible involvement of TRPC channels in the development of contractile dysfunction associated with reperfusion of ischemic myocardium using a mouse heart model. The functional expression of TRPC channels was confirmed in mouse ventricular myocytes using immunocytochemistry, Western blotting, and patch-clamp experiments. The left ventricular functions were assessed by measuring left ventricular end-diastolic pressure, left ventricular developed pressure, and its first derivatives in a Langendorff-perfused mouse heart subjected to 30 minutes of normothermic (37°C) global ischemia followed by 60 minutes of reperfusion. Under control conditions, left ventricular functions were deteriorated during reperfusion, which was significantly ameliorated by administration of the TRPC channel blockers 2-aminoethoxydiphenyl borate and La during initial 5 minutes of reperfusion. Our findings suggest that TRPC channels are involved in mediating contractile dysfunction during reperfusion of ischemic myocardium and detect TRPC channels as a potential therapeutic target for preventing myocardial ischemia/reperfusion injury.
Collapse
|
49
|
Koenig X, Choi RH, Schicker K, Singh DP, Hilber K, Launikonis BS. Mechanistic insights into store-operated Ca 2+ entry during excitation-contraction coupling in skeletal muscle. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1239-1248. [PMID: 30825472 DOI: 10.1016/j.bbamcr.2019.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/11/2023]
Abstract
Skeletal muscle fibres support store-operated Ca2+-entry (SOCE) across the t-tubular membrane upon exhaustive depletion of Ca2+ from the sarcoplasmic reticulum (SR). Recently we demonstrated the presence of a novel mode of SOCE activated under conditions of maintained [Ca2+]SR. This phasic SOCE manifested in a fast and transient manner in synchrony with excitation contraction (EC)-coupling mediated SR Ca2+-release (Communications Biology 1:31, doi: https://doi.org/10.1038/s42003-018-0033-7). Stromal interaction molecule 1 (STIM1) and calcium release-activated calcium channel 1 (ORAI1), positioned at the SR and t-system membranes, respectively, are the considered molecular correlate of SOCE. The evidence suggests that at the triads, where the terminal cisternae of the SR sandwich a t-tubule, STIM1 and ORAI1 proteins pre-position to allow for enhanced SOCE transduction. Here we show that phasic SOCE is not only shaped by global [Ca2+]SR but provide evidence for a local activation within nanodomains at the terminal cisternae of the SR. This feature may allow SOCE to modulate [Ca2+]SR during EC coupling. We define SOCE to occur on the same timescale as EC coupling and determine the temporal coherence of SOCE activation to SR Ca2+ release. We derive a delay of 0.3 ms reflecting diffusive Ca2+-equilibration at the luminal ryanodine receptor 1 (RyR1) channel mouth upon SR Ca2+-release. Numerical simulations of Ca2+-calsequestrin binding estimates a characteristic diffusion length and confines an upper limit for the spatial distance between STIM1 and RyR1. Experimental evidence for a 4- fold change in t-system Ca2+-permeability upon prolonged electrical stimulation in conjunction with numerical simulations of Ca2+-STIM1 binding suggests a Ca2+ dissociation constant of STIM1 below 0.35 mM. Our results show that phasic SOCE is intimately linked with RyR opening and closing, with only μs delays, because [Ca2+] in the terminal cisternae is just above the threshold for Ca2+ dissociation from STIM1 under physiological resting conditions. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Xaver Koenig
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria.
| | - Rocky H Choi
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Klaus Schicker
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria
| | - Daniel P Singh
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria
| | - Bradley S Launikonis
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
50
|
Pollak AJ, Liu C, Gudlur A, Mayfield JE, Dalton ND, Gu Y, Chen J, Heller Brown J, Hogan PG, Wiley SE, Peterson KL, Dixon JE. A secretory pathway kinase regulates sarcoplasmic reticulum Ca 2+ homeostasis and protects against heart failure. eLife 2018; 7:41378. [PMID: 30520731 PMCID: PMC6298778 DOI: 10.7554/elife.41378] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022] Open
Abstract
Ca2+ signaling is important for many cellular and physiological processes, including cardiac function. Although sarcoplasmic reticulum (SR) proteins involved in Ca2+ signaling have been shown to be phosphorylated, the biochemical and physiological roles of protein phosphorylation within the lumen of the SR remain essentially uncharacterized. Our laboratory recently identified an atypical protein kinase, Fam20C, which is uniquely localized to the secretory pathway lumen. Here, we show that Fam20C phosphorylates several SR proteins involved in Ca2+ signaling, including calsequestrin2 and Stim1, whose biochemical activities are dramatically regulated by Fam20C mediated phosphorylation. Notably, phosphorylation of Stim1 by Fam20C enhances Stim1 activation and store-operated Ca2+ entry. Physiologically, mice with Fam20c ablated in cardiomyocytes develop heart failure following either aging or induced pressure overload. We extended these observations to show that non-muscle cells lacking Fam20C display altered ER Ca2+ signaling. Overall, we show that Fam20C plays an overarching role in ER/SR Ca2+ homeostasis and cardiac pathophysiology.
Collapse
Affiliation(s)
- Adam J Pollak
- Department of Pharmacology, University of California, San Diego, San Diego, United States
| | - Canzhao Liu
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Aparna Gudlur
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, San Diego, United States
| | - Joshua E Mayfield
- Department of Pharmacology, University of California, San Diego, San Diego, United States
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Yusu Gu
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Ju Chen
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, San Diego, United States
| | - Patrick G Hogan
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, San Diego, United States.,Program in Immunology, University of California, San Diego, San Diego, United States.,Moores Cancer Center, University of California, San Diego, San Diego, United States
| | - Sandra E Wiley
- Department of Pharmacology, University of California, San Diego, San Diego, United States
| | - Kirk L Peterson
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Jack E Dixon
- Department of Pharmacology, University of California, San Diego, San Diego, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States.,Department of Chemistry and Biochemistry, University of California, San Diego, San Diego, United States
| |
Collapse
|