1
|
Agrawal N, Afzal M, Almalki WH, Ballal S, Sharma GC, Krithiga T, Panigrahi R, Saini S, Ali H, Goyal K, Rana M, Abida Khan. Longevity mechanisms in cardiac aging: exploring calcium dysregulation and senescence. Biogerontology 2025; 26:94. [PMID: 40259024 DOI: 10.1007/s10522-025-10229-8] [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: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025]
Abstract
Cardiac aging is a multistep process that results in a loss of various structural and functional heart abilities, increasing the risk of heart disease. Since its remarkable discovery in the early 1800s, when limestone is heated, calcium's importance has been defined in numerous ways. It can help stiffen shells and bones, function as a reducing agent in chemical reactions, and play a central role in cellular signalling. The movement of calcium ions in and out of cells and between those is referred to as calcium signalling. It influences the binding of the ligand, enzyme activity, electrochemical gradients, and other cellular processes. Calcium signalling is critical for both contraction and relaxation under the sliding filament model of heart muscle. However, with age, the heart undergoes changes that lead to increases in cardiac dysfunction, such as myocardial fibrosis, decreased cardiomyocyte function, and noxious disturbances in calcium homeostasis. Additionally, when cardiac tissues age, cellular senescence, a state of irreversible cell cycle arrest, accumulates and begins to exacerbate tissue inflammation and fibrosis. This review explores the most recent discoveries regarding the role of senescent cell accumulation and calcium signalling perturbances in cardiac aging. Additionally, new treatment strategies are used to reduce aged-related heart dysfunction by targeting senescent cells and modulating calcium homeostasis.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - T Krithiga
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Abida Khan
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
2
|
He ZX, Gao G, Qiao H, Dong GJ, Dan Z, Li YL, Qi YR, Zhang Q, Yuan S, Liu HM, Dong J, Zhao W, Ma LY. Discovery of 1,2,4-Triazole-3-thione Derivatives as Potent and Selective DCN1 Inhibitors for Pathological Cardiac Fibrosis and Remodeling. J Med Chem 2024; 67:18699-18723. [PMID: 39158077 DOI: 10.1021/acs.jmedchem.4c00713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
DCN1, a critical co-E3 ligase during the neddylation process, is overactivated in many diseases, such as cancers, heart failure as well as fibrotic diseases, and has been regarded as a new target for drug development. Herein, we designed and synthesized a new class of 1,2,4-triazole-3-thione-based DCN1 inhibitors based the hit HD1 identified from high-throughput screening and optimized through numerous structure-activity-relationship (SAR) explorations. HD2 (IC50= 2.96 nM) was finally identified and represented a highly potent and selective DCN1 inhibitor with favorable PK properties and low toxicity. Amazingly, HD2 effectively relieved Ang II/TGFβ-induced cardiac fibroblast activation in vitro, and reduced ISO-induced cardiac fibrosis as well as remodeling in vivo, which was linked to the inhibition of cullin 3 neddylation and its substrate Nrf2 accumulation. Our findings unveil a novel 1,2,4-triazole-3-thione-based derivative HD2, which can be recognized as a promising lead compound targeting DCN1 for cardiac fibrosis and remodeling.
Collapse
Affiliation(s)
- Zhang-Xu He
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ge Gao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hui Qiao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guan-Jun Dong
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zengyangzong Dan
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ya-Lan Li
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yu-Ruo Qi
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shuo Yuan
- Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jianzeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- China Meheco Topfond Pharmaceutical Co., Zhumadian 463000, China
- Key Laboratory of Cardiocerebrovascular Drugs, Zhumadian 463000, Henan Province, China
| |
Collapse
|
3
|
Zhang T, Zhang Y, Li S, Ge H, Song Q, Zhang Y, Yang G, Li A. Gentianella acuta-derived Gen-miR-1 suppresses myocardial fibrosis by targeting HAX1/HMG20A/Smads axis to attenuate inflammation in cardiac fibroblasts. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154923. [PMID: 37352750 DOI: 10.1016/j.phymed.2023.154923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/14/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Continuous activation and inflammation of cardiac fibroblasts (CFs) are essential for myocardial fibrosis. Gentianella acuta (Michx.) Hiitonen (G. acuta), that contains xanthones with cardioprotective properties, a typical healthful herb extensively used to treat cardiovascular diseases in Inner Mongolia region of China. However, it remains unknown whether or not G. acuta-derived miRNAs can shield CFs from activation by inflammatory stimulation. Therefore, we tend to investigated the role and core mechanism of G. acuta-derived Gen-miR-1 in regulating fibrosis and inflammation induced by TGF-β1. METHODS An animal model for myocardial infarction was built by subcutaneous injections of ISO and treated with Gen-miR-1 using intragastric administration. The protective effect of Gen-miR-1 on the heart was assessed by pathomorphological analysis of myocardial fibrosis. Using loss- and gain-of-function approaches, Gen-miR-1 regulation of HAX1/HMG20A/Smads axis was investigated by utilizing luciferase assay, Western blot, co-immunoprecipitation, etc. RESULTS: Screened and identified Gen-miR-1 from G. acuta. Gen-miR-1 can enter the mouse body, and markedly inhibit myocardial infarction induced by ISO in mice, as well as suppresses fibrosis in CFs and attenuates the inflammatory response elicited by TGF-β1 in vitro. Gen-miR-1 downregulates HCLS1-related Protein X-1 (HAX1) expression through direct binding to the 3' UTR of HAX1, which in turn relieves HAX1 from promoting the expression of high-mobility group protein 20A (HMG20A), whereas HMG20A downregulation restrains the activation of TGF-β1/Smads signaling pathways, subsequently resulting in a decrease of fibrosis and in facilitating CFs anti-inflammatory effects induced by Gen-miR-1 in the context of CFs activation induced by TGF-β1. CONCLUSIONS Our results first uncovered unique bioactive components in G. acuta and elucidated the molecular mechanism by which G. acuta-derived Gen-miR-1 suppress inflammation and myocardial fibrosis. These findings expand our understanding of G. acuta's therapeutic properties and bioactive constituents. Gen-miR-1-regulated HAX1/HMG20A/Smads axis will be one potential therapeutic target for cardiac remodeling.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Si Li
- Department of Technology, Hebei University of Chinese Medicine, Shijiazhuang, PR China
| | - Hongyao Ge
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Qiuhang Song
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, Hebei, PR China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang, PR China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, Hebei, PR China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang, PR China
| | - Gaoshan Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, Hebei, PR China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang, PR China.
| | - Aiying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, Hebei, PR China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang, PR China.
| |
Collapse
|
4
|
Ernst P, Bidwell PA, Dora M, Thomas DD, Kamdar F. Cardiac calcium regulation in human induced pluripotent stem cell cardiomyocytes: Implications for disease modeling and maturation. Front Cell Dev Biol 2023; 10:986107. [PMID: 36742199 PMCID: PMC9889838 DOI: 10.3389/fcell.2022.986107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) are based on ground-breaking technology that has significantly impacted cardiovascular research. They provide a renewable source of human cardiomyocytes for a variety of applications including in vitro disease modeling and drug toxicity testing. Cardiac calcium regulation plays a critical role in the cardiomyocyte and is often dysregulated in cardiovascular disease. Due to the limited availability of human cardiac tissue, calcium handling and its regulation have most commonly been studied in the context of animal models. hiPSC-CMs can provide unique insights into human physiology and pathophysiology, although a remaining limitation is the relative immaturity of these cells compared to adult cardiomyocytes Therefore, this field is rapidly developing techniques to improve the maturity of hiPSC-CMs, further establishing their place in cardiovascular research. This review briefly covers the basics of cardiomyocyte calcium cycling and hiPSC technology, and will provide a detailed description of our current understanding of calcium in hiPSC-CMs.
Collapse
Affiliation(s)
- Patrick Ernst
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Philip A. Bidwell
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Michaela Dora
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, United States
| | - David D. Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Forum Kamdar
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
5
|
Fischer M, Jakab M, Hirt MN, Werner TR, Engelhardt S, Sarikas A. Identification of hypertrophy-modulating Cullin-RING ubiquitin ligases in primary cardiomyocytes. Front Physiol 2023; 14:1134339. [PMID: 36969608 PMCID: PMC10030680 DOI: 10.3389/fphys.2023.1134339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Cullin-RING ubiquitin ligases (CRL) regulate numerous biological processes in the heart and have been implicated in regulating cardiac hypertrophy. This study aimed to identify novel hypertrophy-modulating CRLs in cardiomyocytes (CM). A functional genomic approach using siRNA-mediated depletion and automated microscopy was employed to screen for cell size-modulating CRLs in neonatal rat CM. Screening hits were confirmed by 3H-isoleucine incorporation. Of 43 targets screened, siRNA-mediated depletion of Fbxo6, Fbxo45, and Fbxl14 resulted in decreased cell size, whereas depletion of Fbxo9, Fbxo25, Fbxo30, Fbxo32, Fbxo33, Cullin1, Roc1, Ddb1, Fbxw4, and Fbxw5 led to a markedly increased cell size under basal conditions. In CM stimulated with phenylephrine (PE), depletion of Fbxo6, Fbxo25, Fbxo33, Fbxo45, and Fbxw4 further augmented PE-induced hypertrophy. As a proof-of-concept, the CRLFbox25 was analysed by transverse aortic constriction (TAC) resulting in a 4.5-fold increase in Fbxo25 protein concentrations compared to control animals. In cell culture, siRNA-mediated depletion of Fbxo25 resulted in a ∼ 37% increase in CM cell size and ∼41% increase in 3H-isoleucine incorporation. Depleting Fbxo25 resulted in upregulation of Anp and Bnp. In summary, we identified 13 novel CRLs as positive or negative regulators of CM hypertrophy. Of these, CRLFbox25 was further characterized, as a potential modulator of cardiac hypertrophy.
Collapse
Affiliation(s)
- Maximillian Fischer
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Correspondence: Maximillian Fischer, ; Antonio Sarikas,
| | - Moritz Jakab
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - Marc N. Hirt
- Institute of Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Tessa R. Werner
- Institute of Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
- Correspondence: Maximillian Fischer, ; Antonio Sarikas,
| |
Collapse
|
6
|
Aberrant PLN-R14del Protein Interactions Intensify SERCA2a Inhibition, Driving Impaired Ca2+ Handling and Arrhythmogenesis. Int J Mol Sci 2022; 23:ijms23136947. [PMID: 35805951 PMCID: PMC9266971 DOI: 10.3390/ijms23136947] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Phospholamban (PLN), a key modulator of Ca2+-homeostasis, inhibits sarcoplasmic reticulum (SR) calcium-ATPase (SERCA2a) and regulates cardiac contractility. The human PLN mutation R14del has been identified in arrhythmogenic cardiomyopathy patients worldwide and is currently extensively investigated. In search of the molecular mechanisms mediating the pathological phenotype, we examined PLN-R14del associations to known PLN-interacting partners. We determined that PLN-R14del interactions to key Ca2+-handling proteins SERCA2a and HS-1-associated protein X-1 (HAX-1) were enhanced, indicating the super-inhibition of SERCA2a’s Ca2+-affinity. Additionally, histidine-rich calcium binding protein (HRC) binding to SERCA2a was increased, suggesting the inhibition of SERCA2a maximal velocity. As phosphorylation relieves the inhibitory effect of PLN on SERCA2a activity, we examined the impact of phosphorylation on the PLN-R14del/SERCA2a interaction. Contrary to PLN-WT, phosphorylation did not affect PLN-R14del binding to SERCA2a, due to a lack of Ser-16 phosphorylation in PLN-R14del. No changes were observed in the subcellular distribution of PLN-R14del or its co-localization to SERCA2a. However, in silico predictions suggest structural perturbations in PLN-R14del that could impact its binding and function. Our findings reveal for the first time that by increased binding to SERCA2a and HAX-1, PLN-R14del acts as an enhanced inhibitor of SERCA2a, causing a cascade of molecular events contributing to impaired Ca2+-homeostasis and arrhythmogenesis. Relieving SERCA2a super-inhibition could offer a promising therapeutic approach for PLN-R14del patients.
Collapse
|
7
|
The Potential of Hsp90 in Targeting Pathological Pathways in Cardiac Diseases. J Pers Med 2021; 11:jpm11121373. [PMID: 34945845 PMCID: PMC8709342 DOI: 10.3390/jpm11121373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone that interacts with up to 10% of the proteome. The extensive involvement in protein folding and regulation of protein stability within cells makes Hsp90 an attractive therapeutic target to correct multiple dysfunctions. Many of the clients of Hsp90 are found in pathways known to be pathogenic in the heart, ranging from transforming growth factor β (TGF-β) and mitogen activated kinase (MAPK) signaling to tumor necrosis factor α (TNFα), Gs and Gq g-protein coupled receptor (GPCR) and calcium (Ca2+) signaling. These pathways can therefore be targeted through modulation of Hsp90 activity. The activity of Hsp90 can be targeted through small-molecule inhibition. Small-molecule inhibitors of Hsp90 have been found to be cardiotoxic in some cases however. In this regard, specific targeting of Hsp90 by modulation of post-translational modifications (PTMs) emerges as an attractive strategy. In this review, we aim to address how Hsp90 functions, where Hsp90 interacts within pathological pathways, and current knowledge of small molecules and PTMs known to modulate Hsp90 activity and their potential as therapeutics in cardiac diseases.
Collapse
|
8
|
Gong H, Wang P, Yu M, Zhu Y, Teng L, Su Y. The Role of the Hematopoietic Cell-Specific Protein 1-Associated Protein X-1 in Human Papillomavirus 16 E2-Induced Human Cervical Squamous Carcinoma Cell Apoptosis via a Mitochondria-Dependent Pathway. Gynecol Obstet Invest 2021; 86:273-282. [PMID: 34175849 DOI: 10.1159/000515532] [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: 06/28/2020] [Accepted: 02/28/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Human papillomavirus 16 (HPV 16) E2 is a transcriptional regulator that plays a key role in regulating a variety of biological responses. Hematopoietic cell-specific protein 1-related protein X-1 (HAX-1) is a mitochondrial membrane protein, and the HAX-1 gene is involved in the occurrence, growth, invasion, and metastasis of various human malignant tumors. The purpose of this study was to investigate the relationships among HPV 16 E2, the role of HAX-1 gene, and the underlying intracellular apoptotic mechanism of human cervical squamous carcinoma cells (C33a and SiHa). METHODS In this study, HAX-1 expression was examined using real-time polymerase chain reaction, Western blot, and immunohistochemical staining analysis. Apoptosis of cells was assessed by flow cytometry. The mitochondrial function was assessed by the mitochondrial copy number, reactive oxygen species (ROS) generation, the mitochondrial membrane potential (ΔΨm), and mitochondrial morphology. RESULTS Our study demonstrated that the expression of the HAX-1 gene was significantly increased in human cervical carcinoma tissues relative to noncancerous cervix tissues. HPV 16 E2 inhibited HAX-1 protein expression. Overexpression of HAX-1 increased the mitochondrial copy number, decreased the production of ROS, and maintained the integrity of the mitochondrial membrane and morphology. So, enhanced expression of the HAX-1 gene could abrogate the HPV 16 E2-induced cell apoptosis. CONCLUSION Therefore, these data support a mechanism that HAX-1 plays a crucial role in HPV 16 E2-induced human cervical squamous carcinoma cell apoptosis in a mitochondrial-dependent manner.
Collapse
Affiliation(s)
- Hui Gong
- Department of Clinical Laboratory, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Ping Wang
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Yu
- Department of Clinical Laboratory, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yu Zhu
- Department of Clinical Laboratory, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Lichen Teng
- Department of Clinical Laboratory, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yajuan Su
- Department of Clinical Laboratory, Cancer Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Dagvadorj J, Mikulska-Ruminska K, Tumurkhuu G, Ratsimandresy RA, Carriere J, Andres AM, Marek-Iannucci S, Song Y, Chen S, Lane M, Dorfleutner A, Gottlieb RA, Stehlik C, Cassel S, Sutterwala FS, Bahar I, Crother TR, Arditi M. Recruitment of pro-IL-1α to mitochondrial cardiolipin, via shared LC3 binding domain, inhibits mitophagy and drives maximal NLRP3 activation. Proc Natl Acad Sci U S A 2021; 118:e2015632118. [PMID: 33361152 PMCID: PMC7817159 DOI: 10.1073/pnas.2015632118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1β release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1β production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jargalsaikhan Dagvadorj
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Karolina Mikulska-Ruminska
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213
- Institute of Physics, Faculty of Physics Astronomy and Informatics, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
| | - Gantsetseg Tumurkhuu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Jessica Carriere
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Allen M Andres
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Stefanie Marek-Iannucci
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Yang Song
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shuang Chen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Malcolm Lane
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Andrea Dorfleutner
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Christian Stehlik
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Suzanne Cassel
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Fayyaz S Sutterwala
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213;
| | - Timothy R Crother
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Moshe Arditi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| |
Collapse
|
10
|
Alogaili F, Chinnarasu S, Jaeschke A, Kranias EG, Hui DY. Hepatic HAX-1 inactivation prevents metabolic diseases by enhancing mitochondrial activity and bile salt export. J Biol Chem 2020; 295:4631-4646. [PMID: 32079675 DOI: 10.1074/jbc.ra119.012361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/04/2020] [Indexed: 12/26/2022] Open
Abstract
Increasing hepatic mitochondrial activity through pyruvate dehydrogenase and elevating enterohepatic bile acid recirculation are promising new approaches for metabolic disease therapy, but neither approach alone can completely ameliorate disease phenotype in high-fat diet-fed mice. This study showed that diet-induced hepatosteatosis, hyperlipidemia, and insulin resistance can be completely prevented in mice with liver-specific HCLS1-associated protein X-1 (HAX-1) inactivation. Mechanistically, we showed that HAX-1 interacts with inositol 1,4,5-trisphosphate receptor-1 (InsP3R1) in the liver, and its absence reduces InsP3R1 levels, thereby improving endoplasmic reticulum-mitochondria calcium homeostasis to prevent excess calcium overload and mitochondrial dysfunction. As a result, HAX-1 ablation activates pyruvate dehydrogenase and increases mitochondria utilization of glucose and fatty acids to prevent hepatosteatosis, hyperlipidemia, and insulin resistance. In contrast to the reduction of InsP3R1 levels, hepatic HAX-1 deficiency increases bile salt exporter protein levels, thereby promoting enterohepatic bile acid recirculation, leading to activation of bile acid-responsive genes in the intestinal ileum to augment insulin sensitivity and of cholesterol transport genes in the liver to suppress hyperlipidemia. The dual mechanisms of increased mitochondrial respiration and enterohepatic bile acid recirculation due to improvement of endoplasmic reticulum-mitochondria calcium homeostasis with hepatic HAX-1 inactivation suggest that this may be a potential therapeutic target for metabolic disease intervention.
Collapse
Affiliation(s)
- Fawzi Alogaili
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sivaprakasam Chinnarasu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| |
Collapse
|
11
|
Tiano F, Amati F, Cherubini F, Morini E, Vancheri C, Maletta S, Fortuni S, Serio D, Quatrana A, Luffarelli R, Benini M, Alfedi G, Panarello L, Rufini A, Toschi N, Frontali M, Romano S, Marcotulli C, Casali C, Gioiosa S, Mariotti C, Mongelli A, Fichera M, Condò I, Novelli G, Testi R, Malisan F. Frataxin deficiency in Friedreich's ataxia is associated with reduced levels of HAX-1, a regulator of cardiomyocyte death and survival. Hum Mol Genet 2020; 29:471-482. [PMID: 31943004 DOI: 10.1093/hmg/ddz306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/31/2022] Open
Abstract
Frataxin deficiency, responsible for Friedreich's ataxia (FRDA), is crucial for cell survival since it critically affects viability of neurons, pancreatic beta cells and cardiomyocytes. In FRDA, the heart is frequently affected with typical manifestation of hypertrophic cardiomyopathy, which can progress to heart failure and cause premature death. A microarray analysis performed on FRDA patient's lymphoblastoid cells stably reconstituted with frataxin, indicated HS-1-associated protein X-1 (HAX-1) as the most significantly upregulated transcript (FC = +2, P < 0.0006). quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR) and western blot analysis performed on (I) HEK293 stably transfected with empty vector compared to wild-type frataxin and (II) lymphoblasts from FRDA patients show that low frataxin mRNA and protein expression correspond to reduced levels of HAX-1. Frataxin overexpression and silencing were also performed in the AC16 human cardiomyocyte cell line. HAX-1 protein levels are indeed regulated through frataxin modulation. Moreover, correlation between frataxin and HAX-1 was further evaluated in peripheral blood mononuclear cells (PBMCs) from FRDA patients and from non-related healthy controls. A regression model for frataxin which included HAX-1, group membership and group* HAX-1 interaction revealed that frataxin and HAX-1 are associated both at mRNA and protein levels. Additionally, a linked expression of FXN, HAX-1 and antioxidant defence proteins MnSOD and Nrf2 was observed both in PBMCs and AC16 cardiomyocytes. Our results suggest that HAX-1 could be considered as a potential biomarker of cardiac disease in FRDA and the evaluation of its expression might provide insights into its pathogenesis as well as improving risk stratification strategies.
Collapse
Affiliation(s)
- Francesca Tiano
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Francesca Amati
- Section of Medical Genetics, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University San Raffaele, 00166 Rome, Italy
| | - Fabio Cherubini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Elena Morini
- Section of Medical Genetics, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Chiara Vancheri
- Section of Medical Genetics, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Sara Maletta
- Section of Medical Genetics, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Silvia Fortuni
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Dario Serio
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Andrea Quatrana
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Riccardo Luffarelli
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Fratagene Therapeutics Srl, 00133 Rome, Italy
| | - Monica Benini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Fratagene Therapeutics Srl, 00133 Rome, Italy
| | - Giulia Alfedi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Fratagene Therapeutics Srl, 00133 Rome, Italy
| | - Luca Panarello
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Alessandra Rufini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Fratagene Therapeutics Srl, 00133 Rome, Italy
| | - Nicola Toschi
- Medical Physics Section, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- A.A. Martinos Center for Biomedical Imaging, Harvard Medical School, Charlestown, MA 02129, USA
| | - Marina Frontali
- CNR Institute of Translational Pharmacology, 00133 Rome, Italy
| | - Silvia Romano
- Neurosciences, Mental Health and Sensory Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| | - Christian Marcotulli
- Department of Medical Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University of Rome, 04100 Latina, Italy
| | - Carlo Casali
- Department of Medical Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University of Rome, 04100 Latina, Italy
| | - Silvia Gioiosa
- SCAI (Super Computing Applications and Innovations) CINECA, 00185 Rome, Italy
| | - Caterina Mariotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Alessia Mongelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Mario Fichera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Giuseppe Novelli
- Section of Medical Genetics, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Neuromed Institute, IRCCS, 86077 Pozzilli, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
- Fratagene Therapeutics Srl, 00133 Rome, Italy
| | - Florence Malisan
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| |
Collapse
|
12
|
Larsen EK, Weber DK, Wang S, Gopinath T, Blackwell DJ, Dalton MP, Robia SL, Gao J, Veglia G. Intrinsically disordered HAX-1 regulates Ca 2+ cycling by interacting with lipid membranes and the phospholamban cytoplasmic region. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183034. [PMID: 31400305 PMCID: PMC6899184 DOI: 10.1016/j.bbamem.2019.183034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/09/2019] [Accepted: 07/30/2019] [Indexed: 01/14/2023]
Abstract
Hematopoietic-substrate-1 associated protein X-1 (HAX-1) is a 279 amino acid protein expressed ubiquitously. In cardiac muscle, HAX-1 was found to modulate the sarcoendoplasmic reticulum calcium ATPase (SERCA) by shifting its apparent Ca2+ affinity (pCa). It has been hypothesized that HAX-1 binds phospholamban (PLN), enhancing its inhibitory function on SERCA. HAX-1 effects are reversed by cAMP-dependent protein kinase A that phosphorylates PLN at Ser16. To date, the molecular mechanisms for HAX-1 regulation of the SERCA/PLN complex are still unknown. Using enzymatic, in cell assays, circular dichroism, and NMR spectroscopy, we found that in the absence of a binding partner HAX-1 is essentially disordered and adopts a partial secondary structure upon interaction with lipid membranes. Also, HAX-1 interacts with the cytoplasmic region of monomeric and pentameric PLN as detected by NMR and in cell FRET assays, respectively. We propose that the regulation of the SERCA/PLN complex by HAX-1 is mediated by its interactions with lipid membranes, adding another layer of control in Ca2+ homeostatic balance in the heart muscle.
Collapse
Affiliation(s)
- Erik K Larsen
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel K Weber
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Songlin Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tata Gopinath
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Michael P Dalton
- Department of Physiology, Loyola University, Maywood, IL 60153, USA
| | - Seth L Robia
- Department of Physiology, Loyola University, Maywood, IL 60153, USA
| | - Jiali Gao
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; School of Chemical Biology and Technology, Beijing University Graduate School, Shenzhen 518055, China
| | - Gianluigi Veglia
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
13
|
Vamecq J, Papegay B, Nuyens V, Boogaerts J, Leo O, Kruys V. Mitochondrial dysfunction, AMPK activation and peroxisomal metabolism: A coherent scenario for non-canonical 3-methylglutaconic acidurias. Biochimie 2019; 168:53-82. [PMID: 31626852 DOI: 10.1016/j.biochi.2019.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
The occurrence of 3-methylglutaconic aciduria (3-MGA) is a well understood phenomenon in leucine oxidation and ketogenesis disorders (primary 3-MGAs). In contrast, its genesis in non-canonical (secondary) 3-MGAs, a growing-up group of disorders encompassing more than a dozen of inherited metabolic diseases, is a mystery still remaining unresolved for three decades. To puzzle out this anthologic problem of metabolism, three clues were considered: (i) the variety of disorders suggests a common cellular target at the cross-road of metabolic and signaling pathways, (ii) the response to leucine loading test only discriminative for primary but not secondary 3-MGAs suggests these latter are disorders of extramitochondrial HMG-CoA metabolism as also attested by their failure to increase 3-hydroxyisovalerate, a mitochondrial metabolite accumulating only in primary 3-MGAs, (iii) the peroxisome is an extramitochondrial site possessing its own pool and displaying metabolism of HMG-CoA, suggesting its possible involvement in producing extramitochondrial 3-methylglutaconate (3-MG). Following these clues provides a unifying common basis to non-canonical 3-MGAs: constitutive mitochondrial dysfunction induces AMPK activation which, by inhibiting early steps in cholesterol and fatty acid syntheses, pipelines cytoplasmic acetyl-CoA to peroxisomes where a rise in HMG-CoA followed by local dehydration and hydrolysis may lead to 3-MGA yield. Additional contributors are considered, notably for 3-MGAs associated with hyperammonemia, and to a lesser extent in CLPB deficiency. Metabolic and signaling itineraries followed by the proposed scenario are essentially sketched, being provided with compelling evidence from the literature coming in their support.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, CHU Lille, Univ Lille, Department of Biochemistry and Molecular Biology, Laboratory of Hormonology, Metabolism-Nutrition & Oncology (HMNO), Center of Biology and Pathology (CBP) Pierre-Marie Degand, CHRU Lille, EA 7364 RADEME, University of North France, Lille, France.
| | - Bérengère Papegay
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Vincent Nuyens
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Jean Boogaerts
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| | - Véronique Kruys
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| |
Collapse
|
14
|
Hakem Zadeh F, Teng ACT, Kuzmanov U, Chambers PJ, Tupling AR, Gramolini AO. AKAP6 and phospholamban colocalize and interact in HEK-293T cells and primary murine cardiomyocytes. Physiol Rep 2019; 7:e14144. [PMID: 31325238 PMCID: PMC6642276 DOI: 10.14814/phy2.14144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/16/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
Phospholamban (PLN) is an important Ca2+ modulator at the sarcoplasmic reticulum (SR) of striated muscles. It physically interacts and inhibits sarcoplasmic reticulum Ca2+ATPase (SERCA2) function, whereas a protein kinase A (PKA)‐dependent phosphorylation at its serine 16 reverses the inhibition. The underlying mechanism of this post‐translational modification, however, remains not fully understood. Using publicly available databases, we identified A‐kinase anchoring protein 6 (AKAP6) as a candidate that might play some roles in PLN phosphorylation. Immunofluorescence showed colocalization between GFP‐AKAP6 and PLN in transfected HEK‐293T cells and cultured mouse neonatal cardiomyocytes (CMNCs). Co‐immunoprecipitation confirmed the functional interaction between AKAP6 and PLN in HEK‐293T and isolated adult rat cardiomyocytes in response to isoproterenol stimulation. Functionally, AKAP6 promoted Ca2+ uptake activity of SERCA1 in cotransfected HEK‐293T cells despite the presence of PLN. These results were further confirmed in adult rat cardiomyocytes. Immunofluorescence showed colocalization of both proteins around the perinuclear region, while protein–protein interaction was corroborated by immunoprecipitation of the nucleus‐enriched fraction of rat hearts. Our findings suggest AKAP6 as a novel interacting partner to PLN in HEK‐293T and murine cardiomyocytes.
Collapse
Affiliation(s)
- Farigol Hakem Zadeh
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario.,Translational Biology and Engineering Program (TBEP), Ted Rogers Centre for Heart Research, Toronto, Ontario
| | - Allen C T Teng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario.,Translational Biology and Engineering Program (TBEP), Ted Rogers Centre for Heart Research, Toronto, Ontario
| | - Uros Kuzmanov
- Translational Biology and Engineering Program (TBEP), Ted Rogers Centre for Heart Research, Toronto, Ontario
| | - Paige J Chambers
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario
| | - Allan R Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario
| | - Anthony O Gramolini
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario.,Translational Biology and Engineering Program (TBEP), Ted Rogers Centre for Heart Research, Toronto, Ontario
| |
Collapse
|
15
|
Li YL, Cai WF, Wang L, Liu GS, Paul C, Jiang L, Wang B, Gao X, Wang Y, Wu SZ. Identification of the Functional Autophagy-Regulatory Domain in HCLS1-Associated Protein X-1 That Resists Against Oxidative Stress. DNA Cell Biol 2018; 37:432-441. [PMID: 29461873 DOI: 10.1089/dna.2017.3873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
HCLS1 Associated Protein X-1 (HAX1) promotes cell survival through attenuation of the damaged signals from endoplasmic reticulum and mitochondria, which are known as prominent intracellular compartments for the autophagic process under stress conditions. This study investigates whether autophagy can be upregulated in response to HAX1 overexpression and identifies the functional motif in HAX1 responsible for the autophagic induction. Autophagosome accumulation, mitochondrial membrane potential (Δψm), and apoptosis were assessed in HEK293 cells post transduction with full-length or truncated HAX1-encoding genes, while empty vector-transduced cells served as control. Upon the oxidative stress, the enhanced autophagy induction was observed in cells overexpressing HAX1, as well as HAX1 truncations that encode peptide segments ranging from amino acids 127-180 (AA127-180). This protective response was further supported by flow cytometry and Western Blot results, in which oxidative stress-induced Δψm dissipation and the programmed cell death were suppressed in HAX1-overexpressing cells, associated with reduced DNA fragmentation and decreased Caspase-9 cleavage. Interestingly, the HAX1-induced autophagy response was abrogated when AA127-180 was removed, compromising the antiapoptotic effects upon oxidative stress. Overall, these data indicate that autophagy induction is involved in HAX1-induced cell protective mechanism, and AA127-180 serves as the functional autophagy-regulatory domain of this antiapoptotic protein.
Collapse
Affiliation(s)
- Ying-Lan Li
- 1 Research Center for High Altitude Medicine, Qinghai University School of Medicine , Xining, China .,2 Qinghai Provincial People's Hospital , Xining, China .,3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Wen-Feng Cai
- 3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Lei Wang
- 3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Guan-Sheng Liu
- 4 Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Christian Paul
- 3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Lin Jiang
- 3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Boyu Wang
- 5 Samaritan Medical Center , Watertown, New York
| | - Xiang Gao
- 1 Research Center for High Altitude Medicine, Qinghai University School of Medicine , Xining, China
| | - Yigang Wang
- 3 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center , Cincinnati, Ohio
| | - Shi-Zheng Wu
- 1 Research Center for High Altitude Medicine, Qinghai University School of Medicine , Xining, China .,2 Qinghai Provincial People's Hospital , Xining, China
| |
Collapse
|
16
|
Endoplasmic reticulum (ER) stress triggers Hax1-dependent mitochondrial apoptotic events in cardiac cells. Apoptosis 2018; 21:1227-1239. [PMID: 27654581 DOI: 10.1007/s10495-016-1286-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiomyocyte apoptosis is a major process in pathogenesis of a number of heart diseases, including ischemic heart diseases and cardiac failure. Ensuring survival of cardiac cells by blocking apoptotic events is an important strategy to improve cardiac function. Although the role of ER disruption in inducing apoptosis has been demonstrated, we do not yet fully understand how it influences the mitochondrial apoptotic machinery in cardiac cell models. Recent investigations have provided evidences that the prosurvival protein HCLS1-associated protein X-1 (Hax1) protein is intimately associated with the pathogenesis of heart disease, mitochondrial biology, and protection from apoptotic cell death. To study the role of Hax1 upon ER stress induction, Hax1 was overexpressed in cardiac cells subjected to ER stress, and cell death parameters as well as mitochondrial alterations were examined. Our results demonstrated that the Hax1 is significantly downregulated in cardiac cells upon ER stress induction. Moreover, overexpression of Hax1 protected from apoptotic events triggered by Tunicamycin-induced ER stress. Upon treatment with Tunicamycin, Hax1 protected from mitochondrial fission, downregulation of mitofusins 1 and 2 (MFN1 and MFN2), loss of mitochondrial membrane potential (∆Ψm), production of reactive oxygen species (ROS) and apoptotic cell death. Taken together, our results suggest that Hax1 inhibits ER stress-induced apoptosis at both the pre- and post-mitochondrial levels. These findings may offer an opportunity to develop new agents that inhibit cell death in the diseased heart.
Collapse
|
17
|
Deng X, Zhao W, Song L, Ying W, Guo X. Pro-apoptotic effect of TRAIL-transfected endothelial progenitor cells on glioma cells. Oncol Lett 2018; 15:5004-5012. [PMID: 29545899 PMCID: PMC5840765 DOI: 10.3892/ol.2018.7977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 01/11/2018] [Indexed: 12/14/2022] Open
Abstract
Glioma is one of the most common aggressive neuroepithelial malignant tumors in the central nervous system. It has a high recurrence rate and poor prognosis, primarily due to the fact that novel therapeutic agents cannot penetrate the blood-brain barrier (BBB). Endothelial progenitor cells (EPCs) have been reported to move across the BBB and access the tumor site. However, whether EPCs expressing the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induce glioma cell apoptosis requires further investigation. In the present study, EPCs were transfected and stably expressed with TRAIL through lentiviral infection. The pro-apoptotic effect of these TRAIL-expressing EPCs on the SHG44 glioma cell line was investigated. The migration ability of TRAIL-expressing EPCs toward SHG44 cells through the Transwell culture system was investigated via a high-content screening assay. The apoptotic rate and the expression of cleaved caspase-8 and −3 in addition to the cleaved poly(ADP-ribose) polymerase in SHG44 cells significantly increased in the TRAIL-overexpressing EPC treatment group compared with the controls. The increased apoptotic rate was reversed using a caspase inhibitor. The findings suggested that the TRAIL-expressing EPCs induced apoptosis in the SHG44 cells by activating the death receptor pathway, indicating that the TRAIL-expressing EPCs may be a useful strategy for glioma treatment.
Collapse
Affiliation(s)
- Xin Deng
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wen Zhao
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China.,Co-innovation Center of Henan for New Drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Laijun Song
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wei Ying
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinbin Guo
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
18
|
Bidwell PA, Liu GS, Nagarajan N, Lam CK, Haghighi K, Gardner G, Cai WF, Zhao W, Mugge L, Vafiadaki E, Sanoudou D, Rubinstein J, Lebeche D, Hajjar R, Sadoshima J, Kranias EG. HAX-1 regulates SERCA2a oxidation and degradation. J Mol Cell Cardiol 2018; 114:220-233. [PMID: 29169992 PMCID: PMC5801168 DOI: 10.1016/j.yjmcc.2017.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/30/2017] [Accepted: 11/19/2017] [Indexed: 01/14/2023]
Abstract
Ischemia/reperfusion injury is associated with contractile dysfunction and increased cardiomyocyte death. Overexpression of the hematopoietic lineage substrate-1-associated protein X-1 (HAX-1) has been shown to protect from cellular injury but the function of endogenous HAX-1 remains obscure due to early lethality of the knockout mouse. Herein we generated a cardiac-specific and inducible HAX-1 deficient model, which uncovered an unexpected role of HAX-1 in regulation of sarco/endoplasmic reticulum Ca-ATPase (SERCA2a) in ischemia/reperfusion injury. Although ablation of HAX-1 in the adult heart elicited no morphological alterations under non-stress conditions, it diminished contractile recovery and increased infarct size upon ischemia/reperfusion injury. These detrimental effects were associated with increased loss of SERCA2a. Enhanced SERCA2a degradation was not due to alterations in calpain and calpastatin levels or calpain activity. Conversely, HAX-1 overexpression improved contractile recovery and maintained SERCA2a levels. The regulatory effects of HAX-1 on SERCA2a degradation were observed at multiple levels, including intact hearts, isolated cardiomyocytes and sarcoplasmic reticulum microsomes. Mechanistically, HAX-1 ablation elicited increased production of reactive oxygen species at the sarco/endoplasic reticulum compartment, resulting in SERCA2a oxidation and a predisposition to its proteolysis. This effect may be mediated by NAPDH oxidase 4 (NOX4), a novel binding partner of HAX-1. Accordingly, NOX inhibition with apocynin abrogated the effects of HAX-1 ablation in hearts subjected to ischemia/reperfusion injury. Taken together, our findings reveal a role of HAX-1 in the regulation of oxidative stress and SERCA2a degradation, implicating its importance in calcium homeostasis and cell survival pathways.
Collapse
Affiliation(s)
- Philip A Bidwell
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Guan-Sheng Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Chi Keung Lam
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - George Gardner
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Wen-Feng Cai
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Wen Zhao
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Luke Mugge
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Despina Sanoudou
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece; 4th Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Jack Rubinstein
- Division of Cardiology, Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Djamel Lebeche
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Roger Hajjar
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA; Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|
19
|
Guo XB, Deng X, Wei Y. Hematopoietic Substrate-1-Associated Protein X-1 Regulates the Proliferation and Apoptosis of Endothelial Progenitor Cells Through Akt Pathway Modulation. Stem Cells 2017; 36:406-419. [PMID: 29139175 DOI: 10.1002/stem.2741] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/05/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Xin-Bin Guo
- Department of Neuro-interventional Radiology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou People's Republic of China
| | - Xin Deng
- Department of Neuro-surgery; The First Affiliated Hospital of Zhengzhou University; Zhengzhou People's Republic of China
| | - Ying Wei
- Department of Neuro-interventional Radiology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou People's Republic of China
| |
Collapse
|
20
|
Bidwell PA, Haghighi K, Kranias EG. The antiapoptotic protein HAX-1 mediates half of phospholamban's inhibitory activity on calcium cycling and contractility in the heart. J Biol Chem 2017; 293:359-367. [PMID: 29150445 DOI: 10.1074/jbc.ra117.000128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/17/2017] [Indexed: 12/25/2022] Open
Abstract
The antiapoptotic protein HAX-1 (HS-associated protein X-1) localizes to sarcoplasmic reticulum (SR) in the heart and interacts with the small membrane protein phospholamban (PLN), inhibiting the cardiac sarco/endoplasmic reticulum calcium ATPase 2a (SERCA2a) in the regulation of overall calcium handling and heart muscle contractility. However, because global HAX-1 deletion causes early lethality, how much endogenous HAX-1 contributes to PLN's inhibitory activity on calcium cycling is unknown. We therefore generated a cardiac-specific and inducible knock-out mouse model. HAX-1 ablation in the adult heart significantly increased contractile parameters and calcium kinetics, associated with increased SR calcium load. These changes occurred without any changes in the protein expression of SERCA2a, PLN, and ryanodine receptor or in the PLN phosphorylation status. The enhanced calcium cycling in the HAX-1-depleted heart was mediated through increases in the calcium affinity of SERCA2a and reduced PLN-SERCA2a binding. Comparison of the HAX-1 deletion-induced stimulatory effects with those elicited by PLN ablation indicated that HAX-1 mediates ∼50% of the PLN-associated inhibitory effects in the heart. Stimulation with the inotropic and lusitropic agent isoproterenol eliminated the differences among wild-type, HAX-1-deficient, and PLN-deficient hearts, and maximally stimulated contractile and calcium kinetic parameters were similar among these three groups. Furthermore, PLN overexpression in the HAX-1-null cardiomyocytes did not elicit any inhibitory effects, indicating that HAX-1 may limit PLN activity. These findings suggest that HAX-1 is a major mediator of PLN's inhibitory activity and a critical gatekeeper of SR calcium cycling and contractility in the heart.
Collapse
Affiliation(s)
- Philip A Bidwell
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Molecular Biology, Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 115 27 Athens, Greece.
| |
Collapse
|
21
|
Deng X, Song L, Wei Y, Guo XB. Analysis of the expression of HAX-1 gene in human glioma. Neurosci Lett 2017; 657:189-193. [PMID: 28751207 DOI: 10.1016/j.neulet.2017.07.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/14/2017] [Accepted: 07/23/2017] [Indexed: 11/30/2022]
Abstract
Glioma, as the most common aggressive malignant tumor in the central nervous system, is still an insurmountable issue in neural diseases. The proliferation and survival mechanism of glioma cells need to be explored further for the development of glioma treatment. Hematopoietic cell-specific protein 1 associated protein X-1 (HAX-1) is well known for its anti-apoptotic effect. It was reported to play an important role in several malignant tumors. However, the effect of HAX-1 in glioma still remains unknown. This study aimed to investigate the expression of HAX-1 in glioma and the correlation between HAX-1 and the clinicopathological characteristics and prognosis of glioma. Quantitative reverse transcription polymerase chain reaction and Western blot analysis showed that HAX-1 was overexpressed in glioma cell lines compared with normal human astrocytes. This trend was confirmed by comparing the expression of HAX-1 in glioma tissues and nontumorous tissues. The study also analyzed the correlation between the expression of HAX-1 and clinicopathological characteristics of glioma and found the expression of HAX-1 to be highly related to the differentiation and World Health Organization stage of glioma tissues. The survival analysis revealed that HAX-1 was an independent prognostic factor. In conclusion, this novel study suggested that the overexpression of HAX-1 might contribute to the malignant progression of glioma.
Collapse
Affiliation(s)
- Xin Deng
- Department of Neuro-surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| | - Laijun Song
- Department of Neuro-surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| | - Ying Wei
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| | - Xin-Bin Guo
- Department of Neuro-Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| |
Collapse
|
22
|
Overexpression of Ubiquinol-Cytochrome c Reductase Core Protein 1 May Protect H9c2 Cardiac Cells by Binding with Zinc. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1314297. [PMID: 28676853 PMCID: PMC5476884 DOI: 10.1155/2017/1314297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/27/2017] [Accepted: 05/10/2017] [Indexed: 11/18/2022]
Abstract
In several recent studies, proteomics analyses suggest that increase of ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) is cardio-protective. However, direct evidence for this effect has not yet been obtained. Thus, the current study aimed to determine this effect and the mechanism underlying this effect. The results showed that overexpression of UQCRC1 protected H9c2 cardiac cells against in vitro simulated ischemia-reperfusion by maintaining mitochondrial membrane potential and suppressing the expression of caspase-3. These protective effects were significantly enhanced by exogenous Zn2+ but completely abolished by Zn2+-selective chelator TPEN. Furthermore, the upregulation of UQCRC1 reduced the concentration of free Zn2+ in mitochondria, whereas the downregulation of UQCRC1 increased the concentration of free Zn2+ in mitochondria. In conclusion, the overexpression of UQCRC1 can protect H9c2 cardiac cells against simulated ischemia/reperfusion, and this cardio-protective effect is likely mediated by zinc binding.
Collapse
|
23
|
Enhanced effect of VEGF165 on L-type calcium currents in guinea-pig cardiac ventricular myocytes. Biomed Pharmacother 2017; 85:697-703. [DOI: 10.1016/j.biopha.2016.11.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/09/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023] Open
|
24
|
Kostmann’s Disease and HCLS1-Associated Protein X-1 (HAX1). J Clin Immunol 2016; 37:117-122. [DOI: 10.1007/s10875-016-0358-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 11/20/2016] [Indexed: 10/20/2022]
|
25
|
Lin YD, Ko MC, Wu ST, Li SF, Hu JF, Lai YJ, Harn HIC, Laio IC, Yeh ML, Yeh HI, Tang MJ, Chang KC, Su FC, Wei EIH, Lee ST, Chen JH, Hoffman AS, Wu WT, Hsieh PCH. A nanopatterned cell-seeded cardiac patch prevents electro-uncoupling and improves the therapeutic efficacy of cardiac repair. Biomater Sci 2016; 2:567-80. [PMID: 26827729 DOI: 10.1039/c3bm60289c] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The heart is an extremely sophisticated organ with nanoscale anisotropic structure, contractility and electro-conductivity; however, few studies have addressed the influence of cardiac anisotropy on cell transplantation for myocardial repair. Here, we hypothesized that a graft's anisotropy of myofiber orientation determines the mechano-electrical characteristics and the therapeutic efficacy. We developed aligned- and random-orientated nanofibrous electrospun patches (aEP and rEP, respectively) with or without seeding of cardiomyocytes (CMs) and endothelial cells (ECs) to test this hypothesis. Atomic force microscopy showed a better beating frequency and amplitude of CMs when cultured on aEP than that from cells cultured on rEP. For the in vivo test, a total of 66 rats were divided into six groups: sham, myocardial infarction (MI), MI + aEP, MI + rEP, MI + CM-EC/aEP and MI + CM-EC/rEP (n ≥ 10 for each group). Implantation of aEP or rEP provided mechanical support and thus retarded functional aggravation at 56 days after MI. Importantly, CM-EC/aEP implantation further improved therapeutic outcomes, while cardiac deterioration occurred on the CM-EC/rEP group. Similar results were shown by hemodynamic and infarct size examination. Another independent in vivo study was performed and electrocardiography and optical mapping demonstrated that there were more ectopic activities and defective electro-coupling after CM-EC/rEP implantation, which worsened cardiac functions. Together these results provide comprehensive functional characterizations and demonstrate the therapeutic efficacy of a nanopatterned anisotropic cardiac patch. Importantly, the study confirms the significance of cardiac anisotropy recapitulation in myocardial tissue engineering, which is valuable for the future development of translational nanomedicine.
Collapse
Affiliation(s)
- Yi-Dong Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan and Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan and Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Surgery, National Cheng Kung University & Hospital, Tainan, Taiwan
| | - Ming-Chin Ko
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan and Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Surgery, National Cheng Kung University & Hospital, Tainan, Taiwan
| | - Su-Ting Wu
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Feng Li
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Jung-Feng Hu
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Jun Lai
- Departments of Internal Medicine and Medical Research, Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan
| | - Hans I-Chen Harn
- Institute of Physiology, National Cheng Kung University, Tainan, Taiwan and Institute of Basic Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Chuang Laio
- Department of Pathology, National Cheng Kung University & Hospital, Tainan, Taiwan
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Hung-I Yeh
- Departments of Internal Medicine and Medical Research, Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan
| | - Ming-Jer Tang
- Institute of Physiology, National Cheng Kung University, Tainan, Taiwan and Institute of Basic Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kung-Chao Chang
- Department of Pathology, National Cheng Kung University & Hospital, Tainan, Taiwan
| | - Fong-Chin Su
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Erika I H Wei
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sho-Tone Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jyh-Hong Chen
- Department of Medicine, National Cheng Kung University & Hospital, Tainan, Taiwan
| | - Allan S Hoffman
- Department of Bioengineering, University of Washington, Seattle, Washington, USA.
| | - Wen-Teng Wu
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan and Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan and Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Surgery, National Cheng Kung University & Hospital, Tainan, Taiwan and Institute of Basic Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Bioengineering, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
26
|
Hirasaka K, Mills EM, Haruna M, Bando A, Ikeda C, Abe T, Kohno S, Nowinski SM, Lago CU, Akagi KI, Tochio H, Ohno A, Teshima-Kondo S, Okumura Y, Nikawa T. UCP3 is associated with Hax-1 in mitochondria in the presence of calcium ion. Biochem Biophys Res Commun 2016; 472:108-13. [PMID: 26915802 DOI: 10.1016/j.bbrc.2016.02.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
Abstract
Uncoupling protein 3 (UCP3) is known to regulate energy dissipation, proton leakage, fatty acid oxidation, and oxidative stress. To identify the putative protein regulators of UCP3, we performed yeast two-hybrid screens. Here we report that UCP3 interacted with HS-1 associated protein X-1 (Hax-1), an anti-apoptotic protein that was localized in the mitochondria, and is involved in cellular responses to Ca(2+). The hydrophilic sequences within loop 2, and the matrix-localized hydrophilic domain of mouse UCP3, were necessary for binding to Hax-1 at the C-terminal domain, adjacent to the mitochondrial inner membrane. Interestingly, interaction of these proteins occurred in a calcium-dependent manner. Moreover, the NMR spectrum of the C-terminal domain of Hax-1 was dramatically changed by removal of Ca(2+), suggesting that the C-terminal domain of Hax-1 underwent a Ca(2+)-induced conformational change. In the Ca(2+)-free state, the C-terminal Hax-1 tended to unfold, suggesting that Ca(2+) binding may induce protein folding of the Hax-1 C-terminus. These results suggested that the UCP3-Hax-1 complex may regulate mitochondrial functional changes caused by mitochondrial Ca(2+).
Collapse
Affiliation(s)
- Katsuya Hirasaka
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, Nagasaki, Japan; Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan.
| | - Edward M Mills
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Marie Haruna
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Aki Bando
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Chika Ikeda
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Tomoki Abe
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Shohei Kohno
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Sara M Nowinski
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Cory U Lago
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ken-Ichi Akagi
- Section of Laboratory Equipment, National Institute of Biomedical Innovation, Osaka, Japan
| | - Hidehito Tochio
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ayako Ohno
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Shigetada Teshima-Kondo
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Yuushi Okumura
- Department of Nutrition and Health, Sagami Woman's University, Kanagawa, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| |
Collapse
|
27
|
Abstract
The various isoforms of the sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA) are responsible for the Ca(2+) uptake from the cytosol into the endoplasmic or sarcoplasmic reticulum (ER/SR). In some tissues, the activity of SERCA can be modulated by binding partners, such as phospholamban and sarcolipin. The activity of SERCA can be characterized by its apparent affinity for Ca(2+) as well as maximal enzymatic velocity. Both parameters can be effectively determined by the protocol described here. Specifically, we describe the measurement of the rate of oxalate-facilitated (45)Ca uptake into the SR of crude mouse ventricular homogenates. This protocol can easily be adapted for different tissues and animal models as well as cultured cells.
Collapse
Affiliation(s)
- Philip A Bidwell
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA.
| |
Collapse
|
28
|
Haghighi K, Pritchard TJ, Liu GS, Singh VP, Bidwell P, Lam CK, Vafiadaki E, Das P, Ma J, Kunduri S, Sanoudou D, Florea S, Vanderbilt E, Wang HS, Rubinstein J, Hajjar RJ, Kranias EG. Human G109E-inhibitor-1 impairs cardiac function and promotes arrhythmias. J Mol Cell Cardiol 2015; 89:349-59. [PMID: 26455482 PMCID: PMC4689614 DOI: 10.1016/j.yjmcc.2015.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/18/2015] [Accepted: 10/04/2015] [Indexed: 01/09/2023]
Abstract
A hallmark of human and experimental heart failure is deficient sarcoplasmic reticulum (SR) Ca-uptake reflecting impaired contractile function. This is at least partially attributed to dephosphorylation of phospholamban by increased protein phosphatase 1 (PP1) activity. Indeed inhibition of PP1 by transgenic overexpression or gene-transfer of constitutively active inhibitor-1 improved Ca-cycling, preserved function and decreased fibrosis in small and large animal models of heart failure, suggesting that inhibitor-1 may represent a potential therapeutic target. We recently identified a novel human polymorphism (G109E) in the inhibitor-1 gene with a frequency of 7% in either normal or heart failure patients. Transgenic mice, harboring cardiac-specific expression of G109E inhibitor-1, exhibited decreases in contractility, Ca-kinetics and SR Ca-load. These depressive effects were relieved by isoproterenol stimulation. Furthermore, stress conditions (2Hz +/- Iso) induced increases in Ca-sparks, Ca-waves (60% of G109E versus 20% in wild types) and after-contractions (76% of G109E versus 23% of wild types) in mutant cardiomyocytes. Similar findings were obtained by acute expression of the G109E variant in adult cardiomyocytes in the absence or presence of endogenous inhibitor-1. The underlying mechanisms included reduced binding of mutant inhibitor-1 to PP1, increased PP1 activity, and dephosphorylation of phospholamban at Ser16 and Thr17. However, phosphorylation of the ryanodine receptor at Ser2808 was not altered while phosphorylation at Ser2814 was increased, consistent with increased activation of Ca/calmodulin-dependent protein kinase II (CaMKII), promoting aberrant SR Ca-release. Parallel in vivo studies revealed that mutant mice developed ventricular ectopy and complex ventricular arrhythmias (including bigeminy, trigeminy and ventricular tachycardia), when challenged with isoproterenol. Inhibition of CaMKII activity by KN-93 prevented the increased propensity to arrhythmias. These findings suggest that the human G109E inhibitor-1 variant impairs SR Ca-cycling and promotes arrhythmogenesis under stress conditions, which may present an additional insult in the compromised function of heart failure carriers.
Collapse
Affiliation(s)
- Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Tracy J Pritchard
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Guan-Sheng Liu
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Vivek P Singh
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Philip Bidwell
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Chi Keung Lam
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece
| | - Parthib Das
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Jianyong Ma
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Swati Kunduri
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Despina Sanoudou
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece; 4th Department of Internal Medicine, Medical School, University of Athens and Attikon Hospital, Greece
| | - Stela Florea
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Erica Vanderbilt
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Hong-Shang Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Jack Rubinstein
- Division of Cardiology, Internal Medicine, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, United States
| | - Roger J Hajjar
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, United States
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States; Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece.
| |
Collapse
|
29
|
Lam CK, Zhao W, Liu GS, Cai WF, Gardner G, Adly G, Kranias EG. HAX-1 regulates cyclophilin-D levels and mitochondria permeability transition pore in the heart. Proc Natl Acad Sci U S A 2015; 112:E6466-75. [PMID: 26553996 PMCID: PMC4664353 DOI: 10.1073/pnas.1508760112] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The major underpinning of massive cell death associated with myocardial infarction involves opening of the mitochondrial permeability transition pore (mPTP), resulting in disruption of mitochondria membrane integrity and programmed necrosis. Studies in human lymphocytes suggested that the hematopoietic-substrate-1 associated protein X-1 (HAX-1) is linked to regulation of mitochondrial membrane function, but its role in controlling mPTP activity remains obscure. Herein we used models with altered HAX-1 expression levels in the heart and uncovered an unexpected role of HAX-1 in regulation of mPTP and cardiomyocyte survival. Cardiac-specific HAX-1 overexpression was associated with resistance against loss of mitochondrial membrane potential, induced by oxidative stress, whereas HAX-1 heterozygous deficiency exacerbated vulnerability. The protective effects of HAX-1 were attributed to specific down-regulation of cyclophilin-D levels leading to reduction in mPTP activation. Accordingly, cyclophilin-D and mPTP were increased in heterozygous hearts, but genetic ablation of cyclophilin-D in these hearts significantly alleviated their susceptibility to ischemia/reperfusion injury. Mechanistically, alterations in cyclophilin-D levels by HAX-1 were contributed by the ubiquitin-proteosomal degradation pathway. HAX-1 overexpression enhanced cyclophilin-D ubiquitination, whereas proteosomal inhibition restored cyclophilin-D levels. The regulatory effects of HAX-1 were mediated through interference of cyclophilin-D binding to heat shock protein-90 (Hsp90) in mitochondria, rendering it susceptible to degradation. Accordingly, enhanced Hsp90 expression in HAX-1 overexpressing cardiomyocytes increased cyclophilin-D levels, as well as mPTP activation upon oxidative stress. Taken together, our findings reveal the role of HAX-1 in regulating cyclophilin-D levels via an Hsp90-dependent mechanism, resulting in protection against activation of mPTP and subsequent cell death responses.
Collapse
Affiliation(s)
- Chi Keung Lam
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - Wen Zhao
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - Guan-Sheng Liu
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - Wen-Feng Cai
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - George Gardner
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - George Adly
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0575
| |
Collapse
|
30
|
SERCA2 Haploinsufficiency in a Mouse Model of Darier Disease Causes a Selective Predisposition to Heart Failure. BIOMED RESEARCH INTERNATIONAL 2015; 2015:251598. [PMID: 26064889 PMCID: PMC4433638 DOI: 10.1155/2015/251598] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/18/2014] [Accepted: 12/23/2014] [Indexed: 12/28/2022]
Abstract
Null mutations in one copy of ATP2A2, the gene encoding sarco/endoplasmic reticulum Ca(2+)-ATPase isoform 2 (SERCA2), cause Darier disease in humans, a skin condition involving keratinocytes. Cardiac function appears to be unimpaired in Darier disease patients, with no evidence that SERCA2 haploinsufficiency itself causes heart disease. However, SERCA2 deficiency is widely considered a contributing factor in heart failure. We therefore analyzed Atp2a2 heterozygous mice to determine whether SERCA2 haploinsufficiency can exacerbate specific heart disease conditions. Despite reduced SERCA2a levels in heart, Atp2a2 heterozygous mice resembled humans in exhibiting normal cardiac physiology. When subjected to hypothyroidism or crossed with a transgenic model of reduced myofibrillar Ca(2+)-sensitivity, SERCA2 deficiency caused no enhancement of the disease state. However, when combined with a transgenic model of increased myofibrillar Ca(2+)-sensitivity, SERCA2 haploinsufficiency caused rapid onset of hypertrophy, decompensation, and death. These effects were associated with reduced expression of the antiapoptotic Hax1, increased levels of the proapoptotic genes Chop and Casp12, and evidence of perturbations in energy metabolism. These data reveal myofibrillar Ca(2+)-sensitivity to be an important determinant of the cardiac effects of SERCA2 haploinsufficiency and raise the possibility that Darier disease patients are more susceptible to heart failure under certain conditions.
Collapse
|
31
|
Haghighi K, Bidwell P, Kranias EG. Phospholamban interactome in cardiac contractility and survival: A new vision of an old friend. J Mol Cell Cardiol 2014; 77:160-7. [PMID: 25451386 PMCID: PMC4312245 DOI: 10.1016/j.yjmcc.2014.10.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/06/2014] [Accepted: 10/09/2014] [Indexed: 01/10/2023]
Abstract
Depressed sarcoplasmic reticulum (SR) calcium cycling, reflecting impaired SR Ca-transport and Ca-release, is a key and universal characteristic of human and experimental heart failure. These SR processes are regulated by multimeric protein complexes, including protein kinases and phosphatases as well as their anchoring and regulatory subunits that fine-tune Ca-handling in specific SR sub-compartments. SR Ca-transport is mediated by the SR Ca-ATPase (SERCA2a) and its regulatory phosphoprotein, phospholamban (PLN). Dephosphorylated PLN is an inhibitor of SERCA2a and phosphorylation by protein kinase A (PKA) or calcium-calmodulin-dependent protein kinases (CAMKII) relieves these inhibitory effects. Recent studies identified additional regulatory proteins, associated with PLN, that control SR Ca-transport. These include the inhibitor-1 (I-1) of protein phosphatase 1 (PP1), the small heat shock protein 20 (Hsp20) and the HS-1 associated protein X-1 (HAX1). In addition, the intra-luminal histidine-rich calcium binding protein (HRC) has been shown to interact with both SERCA2a and triadin. Notably, there is physical and direct interaction between these protein players, mediating a fine-cross talk between SR Ca-uptake, storage and release. Importantly, regulation of SR Ca-cycling by the PLN/SERCA interactome does not only impact cardiomyocyte contractility, but also survival and remodeling. Indeed, naturally occurring variants in these Ca-cycling genes modulate their activity and interactions with other protein partners, resulting in depressed contractility and accelerated remodeling. These genetic variants may serve as potential prognostic or diagnostic markers in cardiac pathophysiology.
Collapse
Affiliation(s)
- Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Philip Bidwell
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
32
|
Di Carlo MN, Said M, Ling H, Valverde CA, De Giusti VC, Sommese L, Palomeque J, Aiello EA, Skapura DG, Rinaldi G, Respress JL, Brown JH, Wehrens XHT, Salas MA, Mattiazzi A. CaMKII-dependent phosphorylation of cardiac ryanodine receptors regulates cell death in cardiac ischemia/reperfusion injury. J Mol Cell Cardiol 2014; 74:274-83. [PMID: 24949568 PMCID: PMC4131282 DOI: 10.1016/j.yjmcc.2014.06.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/22/2014] [Accepted: 06/09/2014] [Indexed: 12/19/2022]
Abstract
Ca(2+)-calmodulin kinase II (CaMKII) activation is deleterious in cardiac ischemia/reperfusion (I/R). Moreover, inhibition of CaMKII-dependent phosphorylations at the sarcoplasmic reticulum (SR) prevents CaMKII-induced I/R damage. However, the downstream targets of CaMKII at the SR level, responsible for this detrimental effect, remain unclear. In the present study we aimed to dissect the role of the two main substrates of CaMKII at the SR level, phospholamban (PLN) and ryanodine receptors (RyR2), in CaMKII-dependent I/R injury. In mouse hearts subjected to global I/R (45/120min), phosphorylation of the primary CaMKII sites, S2814 on cardiac RyR2 and of T17 on PLN, significantly increased at the onset of reperfusion whereas PKA-dependent phosphorylation of RyR2 and PLN did not change. Similar results were obtained in vivo, in mice subjected to regional myocardial I/R (1/24h). Knock-in mice with an inactivated serine 2814 phosphorylation site on RyR2 (S2814A) significantly improved post-ischemic mechanical recovery, reduced infarct size and decreased apoptosis. Conversely, knock-in mice, in which CaMKII site of RyR2 is constitutively activated (S2814D), significantly increased infarct size and exacerbated apoptosis. In S2814A and S2814D mice subjected to regional myocardial ischemia, infarct size was also decreased and increased respectively. Transgenic mice with double-mutant non-phosphorylatable PLN (S16A/T17A) in the PLN knockout background (PLNDM) also showed significantly increased post-ischemic cardiac damage. This effect cannot be attributed to PKA-dependent PLN phosphorylation and was not due to the enhanced L-type Ca(2+) current, present in these mice. Our results reveal a major role for the phosphorylation of S2814 site on RyR2 in CaMKII-dependent I/R cardiac damage. In contrast, they showed that CaMKII-dependent increase in PLN phosphorylation during reperfusion opposes rather than contributes to I/R damage.
Collapse
Affiliation(s)
- Mariano N Di Carlo
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Matilde Said
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Haiyun Ling
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0636, USA
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Verónica C De Giusti
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Leandro Sommese
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Ernesto A Aiello
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Darlene G Skapura
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
| | - Gustavo Rinaldi
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina
| | - Jonathan L Respress
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0636, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
| | - Margarita A Salas
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina.
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, CCT, La Plata, Facultad de Ciencias Médicas, 60 y 120, 1900 La Plata, Argentina.
| |
Collapse
|
33
|
Trebinska A, Högstrand K, Grandien A, Grzybowska EA, Fadeel B. Exploring the anti-apoptotic role of HAX-1 versus BCL-XL
in cytokine-dependent bone marrow-derived cells from mice. FEBS Lett 2014; 588:2921-7. [DOI: 10.1016/j.febslet.2014.05.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 05/07/2014] [Accepted: 05/22/2014] [Indexed: 10/25/2022]
|
34
|
Mattiazzi A, Kranias EG. The role of CaMKII regulation of phospholamban activity in heart disease. Front Pharmacol 2014; 5:5. [PMID: 24550830 PMCID: PMC3913884 DOI: 10.3389/fphar.2014.00005] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/07/2014] [Indexed: 01/06/2023] Open
Abstract
Phospholamban (PLN) is a phosphoprotein in cardiac sarcoplasmic reticulum (SR) that is a reversible regulator of the Ca2+-ATPase (SERCA2a) activity and cardiac contractility. Dephosphorylated PLN inhibits SERCA2a and PLN phosphorylation, at either Ser16 by PKA or Thr17 by Ca2+-calmodulin-dependent protein kinase (CaMKII), reverses this inhibition. Through this mechanism, PLN is a key modulator of SR Ca2+ uptake, Ca2+ load, contractility, and relaxation. PLN phosphorylation is also the main determinant of β1-adrenergic responses in the heart. Although phosphorylation of Thr17 by CaMKII contributes to this effect, its role is subordinate to the PKA-dependent increase in cytosolic Ca2+, necessary to activate CaMKII. Furthermore, the effects of PLN and its phosphorylation on cardiac function are subject to additional regulation by its interacting partners, the anti-apoptotic HAX-1 protein and Gm or the anchoring unit of protein phosphatase 1. Regulation of PLN activity by this multimeric complex becomes even more important in pathological conditions, characterized by aberrant Ca2+-cycling. In this scenario, CaMKII-dependent PLN phosphorylation has been associated with protective effects in both acidosis and ischemia/reperfusion. However, the beneficial effects of increasing SR Ca2+ uptake through PLN phosphorylation may be lost or even become deleterious, when these occur in association with alterations in SR Ca2+ leak. Moreover, a major characteristic in human and experimental heart failure (HF) is depressed SR Ca2+ uptake, associated with decreased SERCA2a levels and dephosphorylation of PLN, leading to decreased SR Ca2+ load and impaired contractility. Thus, the strategy of altering SERCA2a and/or PLN levels or activity to restore perturbed SR Ca2+ uptake is a potential therapeutic tool for HF treatment. We will review here the role of CaMKII-dependent phosphorylation of PLN at Thr17 on cardiac function under physiological and pathological conditions.
Collapse
Affiliation(s)
- Alicia Mattiazzi
- Facultad de Medicina, Centro de Investigaciones Cardiovasculares, Conicet La Plata-Universidad Nacional de La Plata La Plata, Argentina
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati Cincinnati, OH, USA
| |
Collapse
|
35
|
Koontz J, Kontrogianni-Konstantopoulos A. Competition through dimerization between antiapoptotic and proapoptotic HS-1-associated protein X-1 (Hax-1). J Biol Chem 2013; 289:3468-77. [PMID: 24347163 DOI: 10.1074/jbc.m113.536151] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies on Hax-1 have mainly focused on variant (v) 1, demonstrating its antiapoptotic properties. However, HAX1 is heavily spliced, generating structurally distinct isoforms. We sought to characterize the Hax-1 isoforms expressed in rat heart before and after insult. We confirmed the presence of at least four Hax-1 transcripts in healthy rat cardiac muscle. These exhibited differential expression before and after induction of myocardial infarction, with v2 being up-regulated 12-fold at the transcript level and 1.5-fold at the protein level post-insult. Contrary to antiapoptotic rat and human v1, overexpression of rat v2 or human v4 (the human homologue of rat v2) in epithelial cells exacerbated cell death by 30% following H2O2 treatment compared with control vector. Coexpression of rat v1 and v2 or human v1 and v4 neutralized the protective effects of rat and human v1 and the proapoptotic effects of rat v2 and human v4 by modulating cytochrome c release. This is, at least partly, mediated by the ability of Hax-1 proteins to form homotypic and heterotypic dimers with binding affinities ranging from ~3.8 nm for v1 dimers to ~97 nm for v1/v2 dimers. The minimal binding region supporting these interactions lies between amino acids 97-278, which are shared by nearly all Hax-1 proteins, indicating that additional factors regulate the preferential formation of Hax-1 homo- or heterodimers. Our studies are the first to show that Hax-1 is a family of anti- and proapoptotic regulators that may modulate cell survival and death through homo- or heterodimerization.
Collapse
Affiliation(s)
- Jason Koontz
- From the Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | | |
Collapse
|
36
|
Lam AK, Galione A, Lai FA, Zissimopoulos S. Hax-1 identified as a two-pore channel (TPC)-binding protein. FEBS Lett 2013; 587:3782-6. [DOI: 10.1016/j.febslet.2013.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 10/26/2022]
|
37
|
Hu J, Mu C, Hao J. Cerebral ischemia reduces expression of Hs1-associated protein X-1 (Hax-1) in mouse brain. Neurosci Lett 2012; 534:338-43. [PMID: 23262083 DOI: 10.1016/j.neulet.2012.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 11/16/2022]
Abstract
Hax-1, a multi-functional protein, recently was found to be involved in apoptosis and nerve system development. The purpose of this study was to detect the effect of cerebral ischemia on Hax-1 expression. We have detected the expression of Hax-1 in normal brain tissue and in ischemic brain tissue. Hax-1 was expressed in all brain regions detected with a level similar to the level of β-actin. There were no differences in the expression of Hax-1 in different brain regions detected. The confocal images confirmed that neurons expressed Hax-1. The results of ischemic stroke in vivo indicated that Hax-1 level was significantly reduced at 24h after ischemia in the ischemic hemisphere, which was only 37%±4.8 of healthy hemisphere (p<0.05), and there was a strong reverse correlation between the level of Hax-1 and infarct size indicated by the regress analysis (R(2)=0.84). The expression of Hax-1 was also reduced in the cells subjected to oxygen/glucose deprivation (OGD) (p<0.01). The expression of Hax-1 was 87%±4.6, 78%±4.9 and 54%±8.2 of control in the murine brain endothelial cell (bEND5 cell) at 1h, 2h and 16h OGD, respectively. The Hax-1 level was 82%±7.3 and 61%±8.1 of control in neuronal cell line (neuro-2a cells) at 5h and 12h OGD, respectively. The percentage of neuro-2a cell death was 40%±11 induced by a 5h of OGD compared to only 10%±4.2 cell death in the control group (p<0.01). Our present study provides preliminary evidence of the effect of cerebral ischemia on Hax-1 expression. The expression of Hax-1 in normal brain tissue and reduction of Hax-1 in ischemic brain tissue indicate its possible involvement in pathophysiological functions in the brain.
Collapse
Affiliation(s)
- Jing Hu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | |
Collapse
|
38
|
Lam CK, Zhao W, Cai W, Vafiadaki E, Florea SM, Ren X, Liu Y, Robbins N, Zhang Z, Zhou X, Jiang M, Rubinstein J, Jones WK, Kranias EG. Novel role of HAX-1 in ischemic injury protection involvement of heat shock protein 90. Circ Res 2012; 112:79-89. [PMID: 22982986 DOI: 10.1161/circresaha.112.279935] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Ischemic heart disease is characterized by contractile dysfunction and increased cardiomyocyte death, induced by necrosis and apoptosis. Increased cell survival after an ischemic insult is critical and depends on several cellular pathways, which have not been fully elucidated. OBJECTIVE To test the hypothesis that the anti-apoptotic hematopoietic lineage substrate-1-associated protein X-1 (HAX-1), recently identified as regulator of cardiac Ca cycling, also may ameliorate cellular injury with an ischemic insult. METHODS AND RESULTS We report that cardiac ischemia/reperfusion injury is associated with significant decreases in HAX-1 levels ex vivo and in vivo. Accordingly, overexpression of HAX-1 improved contractile recovery, coupled with reduced infarct size, plasma troponin I level, and apoptosis. The beneficial effects were associated with decreased endoplasmic reticulum (ER) stress response through specific inhibition of the inositol-requiring enzyme (IRE-1) signaling pathway, including its downstream effectors caspase-12 and the transcription factor C/EBP homologous protein. Conversely, HAX-1 heterozygous-deficient hearts exhibited increases in infarct size and IRE-1 activity. The inhibitory effects of HAX-1 were mediated by its binding to the N-terminal fragment of the heat shock protein 90 (Hsp90). Moreover, HAX-1 sequestered Hsp90 from IRE-1 to the phospholamban-sarcoplasmic/endoplasmic reticulum calcium ATPase complex. The HAX-1 regulation was further supported by loss of IRE-1 inhibition in presence of the Hsp90 inhibitor, 17-N-allylamino-17-demethoxygeldanamycin. CONCLUSIONS Cardiac ischemia-reperfusion injury is associated with decreases in HAX-1 levels. Consequently, overexpression of HAX-1 promotes cardiomyocyte survival, mediated by its interaction with Hsp90 and specific inhibition of IRE-1 signaling at the ER/sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Chi Keung Lam
- Department of Pharmacology & Cell Biophysics, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Heart disease remains the leading cause of death and disability in the Western world. Current therapies aim at treating the symptoms rather than the subcellular mechanisms, underlying the etiology and pathological remodeling in heart failure. A universal characteristic, contributing to the decreased contractile performance in human and experimental failing hearts, is impaired calcium sequestration into the sarcoplasmic reticulum (SR). SR calcium uptake is mediated by a Ca(2+)-ATPase (SERCA2), whose activity is reversibly regulated by phospholamban (PLN). Dephosphorylated PLN is an inhibitor of SERCA and phosphorylation of PLN relieves this inhibition. However, the initial simple view of a PLN/SERCA regulatory complex has been modified by our recent identification of SUMO, S100 and the histidine-rich Ca-binding protein as regulators of SERCA activity. In addition, PLN activity is regulated by 2 phosphoproteins, the inhibitor-1 of protein phosphatase 1 and the small heat shock protein 20, which affect the overall SERCA-mediated Ca-transport. This review will highlight the regulatory mechanisms of cardiac contractility by the multimeric SERCA/PLN-ensemble and the potential for new therapeutic avenues targeting this complex by using small molecules and gene transfer methods.
Collapse
Affiliation(s)
- Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0575, USA.
| | | |
Collapse
|
40
|
Louridas GE, Lourida KG. A conceptual paradigm of heart failure and systems biology approach. Int J Cardiol 2012; 159:5-13. [DOI: 10.1016/j.ijcard.2011.07.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 07/03/2011] [Indexed: 10/17/2022]
|
41
|
Li B, Hu Q, Xu R, Ren H, Fei E, Chen D, Wang G. Hax-1 is rapidly degraded by the proteasome dependent on its PEST sequence. BMC Cell Biol 2012; 13:20. [PMID: 22827267 PMCID: PMC3432607 DOI: 10.1186/1471-2121-13-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 07/13/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND HS-1-associated protein X-1 (Hax-1), is a multifunctional protein that has sequence homology to Bcl-2 family members. HAX-1 knockout animals reveal that it plays an essential protective role in the central nervous system against various stresses. Homozygous mutations in the HAX-1 gene are associated with autosomal recessive forms of severe congenital neutropenia along with neurological symptoms. The protein level of Hax-1 has been shown to be regulated by cellular protease cleavage or by transcriptional suppression upon stimulation. RESULTS Here, we report a novel post-translational mechanism for regulation of Hax-1 levels in mammalian cells. We identified that PEST sequence, a sequence rich in proline, glutamic acid, serine and threonine, is responsible for its poly-ubiquitination and rapid degradation. Hax-1 is conjugated by K48-linked ubiquitin chains and undergoes a fast turnover by the proteasome system. A deletion mutant of Hax-1 that lacks the PEST sequence is more resistant to the proteasomal degradation and exerts more protective effects against apoptotic stimuli than wild type Hax-1. CONCLUSION Our data indicate that Hax-1 is a short-lived protein and that its PEST sequence dependent fast degradation by the proteasome may contribute to the rapid cellular responses upon different stimulations.
Collapse
Affiliation(s)
- Bin Li
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Soochow University College of Pharmaceutical Sciences, Suzhou, Jiangsu, 201203, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
42
|
Mazzio EA, Boukli N, Rivera N, Soliman KFA. Pericellular pH homeostasis is a primary function of the Warburg effect: inversion of metabolic systems to control lactate steady state in tumor cells. Cancer Sci 2012; 103:422-32. [PMID: 22320183 DOI: 10.1111/j.1349-7006.2012.02206.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 11/22/2011] [Accepted: 12/08/2011] [Indexed: 12/25/2022] Open
Abstract
The Warburg effect describes a heightened propensity of tumor cells to produce lactic acid in the presence or absence of O(2) . A generally held notion is that the Warburg effect is related to energy. Using whole-genome, proteomic MALDI-TOF-MS and metabolite analysis, we investigated the Warburg effect in malignant neuroblastoma N2a cells. The findings show that the Warburg effect serves a functional role in regulating acidic pericellular pH (pHe), which is mediated by metabolic inversion or a fluctuating dominance between glycolytic-rate substrate level phosphorylation (SLP) and mitochondrial (mt) oxidative phosphorylation (OXPHOS) to control lactic acid production. The results also show that an alkaline pHe caused an elevation in SLP/OXPHOS ratio (approximately 98% SLP/OXPHOS); while the ratio was approximately 56% at neutral pHe and approximately 93% in acidic pHe. Acidic pHe paralleled greater expression of mitochondrial biogenesis and OXPHOS genes, such as complex III-V (Uqcr10, Atp5 and Cox7c), mt Fmc1, Romo1, Tmem 173, Tomm6, aldehyde dehydrogenase, mt Sod2 mt biogenesis component PPAR-γ co-activator 1 adjunct to loss of mt fission (Mff). Moreover, acidic pHe corresponded to metabolic efficiency evidenced by a rise in mTOR nutrient sensor GβL, its downstream target (Eif4ebp1), insulin modulators (Trib3 and Fetub) and loss of catabolic (Hadhb, Bdh1 and Pygl)/glycolytic processes (aldolase C, pyruvate kinase, Nampt and aldose-reductase). In contrast, alkaline pHe initiated loss of mitofusin 2, complex II-IV (Sdhaf1, Uqcrq, Cox4i2 and Aldh1l2), aconitase, mitochondrial carrier triple repeat 1 and mt biosynthetic (Coq2, Coq5 and Coq9). In conclusion, the Warburg effect might serve as a negative feedback loop that regulates the pHe toward a broad acidic range by altering lactic acid production through inversion of metabolic systems. These effects were independent of changes in O(2) concentration or glucose supply.
Collapse
Affiliation(s)
- Elizabeth A Mazzio
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, Florida, USA
| | | | | | | |
Collapse
|
43
|
Yap SV, Koontz JM, Kontrogianni-Konstantopoulos A. HAX-1: A family of apoptotic regulators in health and disease. J Cell Physiol 2011; 226:2752-61. [DOI: 10.1002/jcp.22638] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
44
|
Klein C. Genetic defects in severe congenital neutropenia: emerging insights into life and death of human neutrophil granulocytes. Annu Rev Immunol 2011; 29:399-413. [PMID: 21219176 DOI: 10.1146/annurev-immunol-030409-101259] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of genetic defects causing congenital neutropenia has illuminated mechanisms controlling differentiation, circulation, and decay of neutrophil granulocytes. Deficiency of the mitochondrial proteins HAX1 and AK2 cause premature apoptosis of myeloid progenitor cells associated with dissipation of the mitochondrial membrane potential, whereas mutations in ELA2/ELANE and G6PC3 are associated with signs of increased endoplasmic reticulum stress. Mutations in the transcriptional repressor GFI1 and the cytoskeletal regulator WASP also lead to defective neutrophil production. This unexpected diversity of factors suggests that multiple pathways are involved in the pathogenesis of congenital neutropenia.
Collapse
Affiliation(s)
- Christoph Klein
- Department of Pediatric Hematology/Oncology, Hannover Medical School, Germany.
| |
Collapse
|
45
|
Vandecaetsbeek I, Vangheluwe P, Raeymaekers L, Wuytack F, Vanoevelen J. The Ca2+ pumps of the endoplasmic reticulum and Golgi apparatus. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004184. [PMID: 21441596 DOI: 10.1101/cshperspect.a004184] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The various splice variants of the three SERCA- and the two SPCA-pump genes in higher vertebrates encode P-type ATPases of the P(2A) group found respectively in the membranes of the endoplasmic reticulum and the secretory pathway. Of these, SERCA2b and SPCA1a represent the housekeeping isoforms. The SERCA2b form is characterized by a luminal carboxy terminus imposing a higher affinity for cytosolic Ca(2+) compared to the other SERCAs. This is mediated by intramembrane and luminal interactions of this extension with the pump. Other known affinity modulators like phospholamban and sarcolipin decrease the affinity for Ca(2+). The number of proteins reported to interact with SERCA is rapidly growing. Here, we limit the discussion to those for which the interaction site with the ATPase is specified: HAX-1, calumenin, histidine-rich Ca(2+)-binding protein, and indirectly calreticulin, calnexin, and ERp57. The role of the phylogenetically older and structurally simpler SPCAs as transporters of Ca(2+), but also of Mn(2+), is also addressed.
Collapse
Affiliation(s)
- Ilse Vandecaetsbeek
- Laboratory of Ca-transport ATPases, Department of Molecular Cell Biology, K.U. Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
46
|
Simmen T. Hax-1: a regulator of calcium signaling and apoptosis progression with multiple roles in human disease. Expert Opin Ther Targets 2011; 15:741-51. [DOI: 10.1517/14728222.2011.561787] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
47
|
In vivo therapy of myocardial infarction with mesenchymal stem cells modified with prostaglandin I synthase gene improves cardiac performance in mice. Life Sci 2011; 88:455-64. [PMID: 21219910 DOI: 10.1016/j.lfs.2010.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 11/28/2010] [Accepted: 12/15/2010] [Indexed: 01/01/2023]
Abstract
AIM Intra-myocardial injection of adult bone marrow-derived stem cells (MSC) has recently been proposed as a therapy to repair damaged cardiomyocytes after acute myocardial infarction (AMI). PGI(2) has vasodilatation effects; however, the effects of combining both MSC and PGI(2) therapy on AMI have never been evaluated. MAIN METHODS We genetically enhanced prostaglandin I synthase (PGIS) gene expression in mouse mesenchymal stem cells (MSC) using lentiviral vector transduction (MSC(PGIS)). Mice were subjected to an AMI model and injected (intra-myocardially) with either 5×10(4) MSCs or MSC(PGIS) before surgery. Fourteen days post AMI, mice were analyzed with echocardiography, immunohistochemistry, and apoptotic, and traditional tissue assays. KEY FINDINGS Lenti-PGIS transduction did not change any characteristic of the MSCs. PGIS over-expressed MSCs secreted 6-keto-PGF1α in the culture medium and decreased free radical damage during hypoxia/re-oxygenation and H(2)O(2) treatment. Furthermore, splenocyte proliferation was significantly suppressed with MSC(PGIS) as compared with MSCs alone. Fourteen days post AMI, echocardiography showed more improvement in cardiac function of the MSC(PGIS) group than the MSC alone group, sham-operated group, or artery ligation only group. The histology of MSC(PGIS) treated hearts revealed MSCs in the infarcted region and decreased myocardial fibrosis/apoptosis with limited cardiac remodeling. Furthermore, the level of the vascular endothelial growth factor was elevated in the MSC(PGIS) group as compared to the other three groups. SIGNIFICANCE In summary, our results provide both in vitro and in vivo evidence for the beneficial role of MSC(PGIS) in limiting the process of detrimental cardiac remodeling in a mouse AMI model during early stages of the disease.
Collapse
|
48
|
Prasad AM, Inesi G. Silencing calcineurin A subunit reduces SERCA2 expression in cardiac myocytes. Am J Physiol Heart Circ Physiol 2010; 300:H173-80. [PMID: 21057045 DOI: 10.1152/ajpheart.00841.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Resting intracellular Ca(2+) can be raised, in neonatal rat cardiac myocytes, by exposure to very low concentration of thapsigargin (TG). Such a Ca(2+) rise yields calcineurin (CN) activation demonstrated by increased expression of transfected luciferase cDNA under control of nuclear factor of activated T-cells (NFAT) promoter and increased translocation of NFAT to nuclei. We found that exposure of cardiac myocytes to TG is followed by increase of sarcroplasmic reticulum Ca(2+) transport ATPase (SERCA2) expression, which is further increased when CN inactivation by CAMKII (calmodulin-dependent kinase) is prevented with KN93 (CAMKII inhibitor). On the other hand, SERCA2 expression is reduced by CN inhibition with cyclosporine. We have now induced calcineurin A (CNA) α- or β-subunit gene silencing with small interfering RNA (siRNA) and observed strong interference with expression of SERCA2, both in control myocytes and following exposure to TG. Such interference is also obtained following NFAT displacement from CN with 9,10-dihydro-9,10[1',2']-benzenoanthracene-1,4-dione (INCA-6). We have also observed analogous effects on expression of phospholamban (PLB) and Na(+)/Ca(2+) exchanger (NCX). Pertinent to these findings, we have identified, by in-silico analysis, NFAT binding sites in SERCA2, PLB, and NCX1 promoters. Our experiments indicate that activation of the calcineurin-NFAT pathway by rise of resting cytosolic Ca(2+) elevates transcription/expression of SERCA2, PLB, and NCX1, providing a homeostatic mechanism for long-term control of cytosolic Ca(2+).
Collapse
Affiliation(s)
- Anand Mohan Prasad
- California Pacific Medical Center Research Institute, San Francisco, California 94107, USA
| | | |
Collapse
|
49
|
Hill RB, Pellegrini L. The PARL family of mitochondrial rhomboid proteases. Semin Cell Dev Biol 2010; 21:582-92. [PMID: 20045481 DOI: 10.1016/j.semcdb.2009.12.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/10/2009] [Accepted: 12/18/2009] [Indexed: 10/20/2022]
Abstract
Rhomboids are an ancient and conserved family of intramembrane-cleaving proteases, a small group of proteolytic enzymes capable of hydrolyzing a peptide bond within a transmembrane helix that anchors a substrate protein to the membrane. Mitochondrial rhomboids evolved in eukaryotes to coordinate a critical aspect of cell biology, the regulation of mitochondrial membranes dynamics. This function appears to have required the emergence of a structural feature that is unique among all other rhomboids: an additional transmembrane helix (TMH) positioned at the N-terminus of six TMHs that form the core proteolytic domain of all prokaryotic and eukaryotic rhomboids. This "1+6" structure, which is shared only among mitochondrial rhomboids, defines a subfamily of rhomboids with the prototypical family member being mammalian Parl. Here, we present the findings that in 11 years have elevated mitochondrial rhomboids as the gatekeepers of mitochondrial dynamics and apoptosis; further, we discuss the aspects of their biology that are bound to introduce new paradigm shifts in our understanding of how the organelle uses this unique type of protease to govern stress, signaling to the nucleus, and other key mitochondrial activities in health and disease.
Collapse
Affiliation(s)
- R Blake Hill
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| | | |
Collapse
|