1
|
Huang KY, Yu HC, Lu MC, Tseng HYH, Shen JJ, Lin CY, Chen PC, Shen YT, Chung PR, Tsai HK, Zhou SR, Wang CL, Lai NS, Lin TH, Huang HB. Identification of a novel Eps 15 homology domain-containing protein 1 (EHD1) and EHD4-binding motif in phostensin. J Biochem 2025; 177:297-304. [PMID: 39776131 DOI: 10.1093/jb/mvaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Phostensin (PTS) encoded by KIAA1949 binds to protein phosphatase 1, F-actin, Eps 15 homology domain-containing protein 1 (EHD1) and EHD4. Most EHD-binding proteins contain a consensus motif, Asn-Pro-Phe (NPF), which interacts with the C-terminal EH domain of EHD proteins. Nevertheless, the NPF motif is absent in PTS. The binding motif for PTS to interact with EHD1 (or EHD4) remains unknown. Here, we identified that PTS-α binds to EHD1 (or EHD4) through the region of residues 51-80, which contains a consensus motif, 64ILV(X)4(L/V)RL74S. This novel consensus motif is also found in vacuolar protein sorting-35 (vps35). Replacement of 64ILV(X)4(L/V)RL74S with 64AAA(X)4(L/V)RL74S or with 64ILV(X)4AEA74A significantly reduces the binding efficiency of PTS-α to either EHD1 or EHD4 in GST pull-down assay and far western blotting assay. In addition, replacement of 218ILV(X)4VRL228S with 218AAA(X)4AEA228A decreases the binding ability of vps35 to EHD4 in far western blotting assay. Overexpression of the PTS-β in 293 T cells attenuated the endocytic trafficking of transferrin. However, this attenuation of transferrin in endocytic trafficking was disrupted when 293 T cells overexpressed the mutant PTS-β with a defective EHD-binding motif, suggesting that PTS-β can regulate the endocytic recycling via associating with EHD1 or EHD4.
Collapse
Affiliation(s)
- Kuang-Yung Huang
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
| | - Hui-Chun Yu
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Hsien-Yu Huang Tseng
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Jyun-Jie Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Ying Lin
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pin-Chen Chen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Ya-Ting Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pei-Rong Chung
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Hsiao-Kuei Tsai
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Si-Ru Zhou
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Lin Wang
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Ning-Sheng Lai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ta-Hsien Lin
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Sec. 2, Linong St., Taipei 11221, Taiwan
| | - Hsien-Bin Huang
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| |
Collapse
|
2
|
Ayagama T, Charles PD, Bose SJ, Boland B, Priestman DA, Aston D, Berridge G, Fischer R, Cribbs AP, Song Q, Mirams GR, Amponsah K, Heather L, Galione A, Herring N, Kramer H, Capel RA, Platt FM, Schotten U, Verheule S, Burton RA. Compartmentalization proteomics revealed endolysosomal protein network changes in a goat model of atrial fibrillation. iScience 2024; 27:109609. [PMID: 38827406 PMCID: PMC11141153 DOI: 10.1016/j.isci.2024.109609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 06/04/2024] Open
Abstract
Endolysosomes (EL) are known for their role in regulating both intracellular trafficking and proteostasis. EL facilitate the elimination of damaged membranes, protein aggregates, membranous organelles and play an important role in calcium signaling. The specific role of EL in cardiac atrial fibrillation (AF) is not well understood. We isolated atrial EL organelles from AF goat biopsies and conducted a comprehensive integrated omics analysis to study the EL-specific proteins and pathways. We also performed electron tomography, protein and enzyme assays on these biopsies. Our results revealed the upregulation of the AMPK pathway and the expression of EL-specific proteins that were not found in whole tissue lysates, including GAA, DYNLRB1, CLTB, SIRT3, CCT2, and muscle-specific HSPB2. We also observed structural anomalies, such as autophagic-vacuole formation, irregularly shaped mitochondria, and glycogen deposition. Our results provide molecular information suggesting EL play a role in AF disease process over extended time frames.
Collapse
Affiliation(s)
- Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | - Daniel Aston
- Department of Anaesthesia and Critical Care, Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge CB2 0AY, UK
| | | | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - Adam P. Cribbs
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Headington OX3 7LD, UK
| | - Qianqian Song
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kwabena Amponsah
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Lisa Heather
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Holger Kramer
- Mass spectrometry Facility, The MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | | | - Ulrich Schotten
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Sander Verheule
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rebecca A.B. Burton
- Department of Pharmacology, University of Oxford, Oxford, UK
- University of Liverpool, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| |
Collapse
|
3
|
Zhu G, Zhang H, Xia M, Liu Y, Li M. EH domain-containing protein 2 (EHD2): Overview, biological function, and therapeutic potential. Cell Biochem Funct 2024; 42:e4016. [PMID: 38613224 DOI: 10.1002/cbf.4016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
EH domain-containing protein 2 (EHD2) is a member of the EHD protein family and is mainly located in the plasma membrane, but can also be found in the cytoplasm and endosomes. EHD2 is also a nuclear-cytoplasmic shuttle protein. After entering the cell nuclear, EHD2 acts as a corepressor of transcription to inhibit gene transcription. EHD2 regulates a series of biological processes. As a key regulator of endocytic transport, EHD2 is involved in the formation and maintenance of endosomal tubules and vesicles, which are critical for the intracellular transport of proteins and other substances. The N-terminal of EHD2 is attached to the cell membrane, while its C-terminal binds to the actin-binding protein. After binding, EHD2 connects with the actin cytoskeleton, forming the curvature of the membrane and promoting cell endocytosis. EHD2 is also associated with membrane protein trafficking and receptor signaling, as well as in glucose metabolism and lipid metabolism. In this review, we highlight the recent advances in the function of EHD2 in various cellular processes and its potential implications in human diseases such as cancer and metabolic disease. We also discussed the prospects for the future of EHD2. EHD2 has a broad prospect as a therapeutic target for a variety of diseases. Further research is needed to explore its mechanism, which could pave the way for the development of targeted treatments.
Collapse
Affiliation(s)
- Guoqiang Zhu
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hu Zhang
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Min Xia
- Hengyang Medical School, Institute of Clinical Medicine, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Hengyang Medical School, Cancer Research Institute, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yiqi Liu
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mingyong Li
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
4
|
Viggars MR, Sutherland H, Lanmüller H, Schmoll M, Bijak M, Jarvis JC. Adaptation of the transcriptional response to resistance exercise over 4 weeks of daily training. FASEB J 2023; 37:e22686. [PMID: 36468768 DOI: 10.1096/fj.202201418r] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
We present the time course of change in the muscle transcriptome 1 h after the last exercise bout of a daily resistance training program lasting 2, 10, 20, or 30 days. Daily exercise in rat tibialis anterior muscles (5 sets of 10 repetitions over 20 min) induced progressive muscle growth that approached a new stable state after 30 days. The acute transcriptional response changed along with progressive adaptation of the muscle phenotype. For example, expression of type 2B myosin was silenced. Time courses recently synthesized from human exercise studies do not demonstrate so clearly the interplay between the acute exercise response and the longer-term consequences of repeated exercise. We highlight classes of transcripts and transcription factors whose expression increases during the growth phase and declines again as the muscle adapts to a new daily pattern of activity and reduces its rate of growth. Myc appears to play a central role.
Collapse
Affiliation(s)
- Mark R Viggars
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK.,Department of Physiology and Aging, University of Florida, Gainesville, Florida, USA.,Myology Institute, University of Florida, Gainesville, Florida, USA
| | - Hazel Sutherland
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Hermann Lanmüller
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Martin Schmoll
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Manfred Bijak
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Jonathan C Jarvis
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
5
|
York NS, Sanchez-Arias JC, McAdam ACH, Rivera JE, Arbour LT, Swayne LA. Mechanisms underlying the role of ankyrin-B in cardiac and neurological health and disease. Front Cardiovasc Med 2022; 9:964675. [PMID: 35990955 PMCID: PMC9386378 DOI: 10.3389/fcvm.2022.964675] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The ANK2 gene encodes for ankyrin-B (ANKB), one of 3 members of the ankyrin family of proteins, whose name is derived from the Greek word for anchor. ANKB was originally identified in the brain (B denotes “brain”) but has become most widely known for its role in cardiomyocytes as a scaffolding protein for ion channels and transporters, as well as an interacting protein for structural and signaling proteins. Certain loss-of-function ANK2 variants are associated with a primarily cardiac-presenting autosomal-dominant condition with incomplete penetrance and variable expressivity characterized by a predisposition to supraventricular and ventricular arrhythmias, arrhythmogenic cardiomyopathy, congenital and adult-onset structural heart disease, and sudden death. Another independent group of ANK2 variants are associated with increased risk for distinct neurological phenotypes, including epilepsy and autism spectrum disorders. The mechanisms underlying ANKB's roles in cells in health and disease are not fully understood; however, several clues from a range of molecular and cell biological studies have emerged. Notably, ANKB exhibits several isoforms that have different cell-type–, tissue–, and developmental stage– expression profiles. Given the conservation within ankyrins across evolution, model organism studies have enabled the discovery of several ankyrin roles that could shed important light on ANKB protein-protein interactions in heart and brain cells related to the regulation of cellular polarity, organization, calcium homeostasis, and glucose and fat metabolism. Along with this accumulation of evidence suggesting a diversity of important ANKB cellular functions, there is an on-going debate on the role of ANKB in disease. We currently have limited understanding of how these cellular functions link to disease risk. To this end, this review will examine evidence for the cellular roles of ANKB and the potential contribution of ANKB functional variants to disease risk and presentation. This contribution will highlight the impact of ANKB dysfunction on cardiac and neuronal cells and the significance of understanding the role of ANKB variants in disease.
Collapse
Affiliation(s)
- Nicole S. York
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Alexa C. H. McAdam
- Department of Medical Genetics, University of British Columbia, Victoria, BC, Canada
| | - Joel E. Rivera
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Laura T. Arbour
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Victoria, BC, Canada
- *Correspondence: Laura T. Arbour
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Cellular and Physiological Sciences and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Leigh Anne Swayne
| |
Collapse
|
6
|
Ayagama T, Bose SJ, Capel RA, Priestman DA, Berridge G, Fischer R, Galione A, Platt FM, Kramer H, Burton RA. A modified density gradient proteomic-based method to analyze endolysosomal proteins in cardiac tissue. iScience 2021; 24:102949. [PMID: 34466782 PMCID: PMC8384914 DOI: 10.1016/j.isci.2021.102949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/04/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
The importance of lysosomes in cardiac physiology and pathology is well established, and evidence for roles in calcium signaling is emerging. We describe a label-free proteomics method suitable for small cardiac tissue biopsies based on density-separated fractionation, which allows study of endolysosomal (EL) proteins. Density gradient fractions corresponding to tissue lysate; sarcoplasmic reticulum (SR), mitochondria (Mito) (1.3 g/mL); and EL with negligible contamination from SR or Mito (1.04 g/mL) were analyzed using Western blot, enzyme activity assay, and liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis (adapted discontinuous Percoll and sucrose differential density gradient). Kyoto Encyclopedia of Genes and Genomes, Reactome, Panther, and Gene Ontology pathway analysis showed good coverage of RAB proteins and lysosomal cathepsins (including cardiac-specific cathepsin D) in the purified EL fraction. Significant EL proteins recovered included catalytic activity proteins. We thus present a comprehensive protocol and data set of guinea pig atrial EL organelle proteomics using techniques also applicable for non-cardiac tissue.
Collapse
Affiliation(s)
- Thamali Ayagama
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Samuel J. Bose
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Rebecca A. Capel
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | | | - Georgina Berridge
- Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Antony Galione
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Frances M. Platt
- University of Oxford, Department of Pharmacology, Oxford, OX1 3QT UK
| | - Holger Kramer
- Biological Mass Spectrometry and Proteomics Facility, MRC London Institute of Medical Sciences, Imperial College London, London, W12 0NN UK
| | | |
Collapse
|
7
|
Wang X, Chen X, Dobrev D, Li N. The crosstalk between cardiomyocyte calcium and inflammasome signaling pathways in atrial fibrillation. Pflugers Arch 2021; 473:389-405. [PMID: 33511453 DOI: 10.1007/s00424-021-02515-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) is the most frequent arrhythmia in adults. The prevalence and incidence of AF is going to increase substantially over the next few decades. Because AF increases the risk of stroke, heart failure, dementia, and others, it severely impacts the quality of life, morbidity, and mortality. Although the pathogenesis of AF is multifaceted and complex, focal ectopic activity and reentry are considered as the fundamental proarrhythmic mechanisms underlying AF development. Over the past 2 decades, large amount of evidence points to the key role of intracellular Ca2+ dysregulation in both initiation and maintenance of AF. More recently, emerging evidence reveal that NLRP3 (NACHT, LRR, PYD domain-containing 3) inflammasome pathway contributes to the substrate of both triggered activity and reentry, ultimately promoting AF. In this article, we review the current state of knowledge on Ca2+ signaling and NLRP3 inflammasome activity in AF. We also discuss the potential crosstalk between these two quintessential contributors to AF promotion.
Collapse
Affiliation(s)
- Xiaolei Wang
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Xiaohui Chen
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA. .,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Wang L, Ma H, Huang P, Xie Y, Near D, Wang H, Xu J, Yang Y, Xu Y, Garbutt T, Zhou Y, Liu Z, Yin C, Bressan M, Taylor JM, Liu J, Qian L. Down-regulation of Beclin1 promotes direct cardiac reprogramming. Sci Transl Med 2020; 12:eaay7856. [PMID: 33087505 PMCID: PMC8188650 DOI: 10.1126/scitranslmed.aay7856] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 05/07/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
Abstract
Direct reprogramming of fibroblasts to alternative cell fates by forced expression of transcription factors offers a platform to explore fundamental molecular events governing cell fate identity. The discovery and study of induced cardiomyocytes (iCMs) not only provides alternative therapeutic strategies for heart disease but also sheds lights on basic biology underlying CM fate determination. The iCM field has primarily focused on early transcriptome and epigenome repatterning, whereas little is known about how reprogramming iCMs remodel, erase, and exit the initial fibroblast lineage to acquire final cell identity. Here, we show that autophagy-related 5 (Atg5)-dependent autophagy, an evolutionarily conserved self-digestion process, was induced and required for iCM reprogramming. Unexpectedly, the autophagic factor Beclin1 (Becn1) was found to suppress iCM induction in an autophagy-independent manner. Depletion of Becn1 resulted in improved iCM induction from both murine and human fibroblasts. In a mouse genetic model, Becn1 haploinsufficiency further enhanced reprogramming factor-mediated heart function recovery and scar size reduction after myocardial infarction. Mechanistically, loss of Becn1 up-regulated Lef1 and down-regulated Wnt inhibitors, leading to activation of the canonical Wnt/β-catenin signaling pathway. In addition, Becn1 physically interacts with other classical class III phosphatidylinositol 3-kinase (PI3K III) complex components, the knockdown of which phenocopied Becn1 depletion in cardiac reprogramming. Collectively, our study revealed an inductive role of Atg5-dependent autophagy as well as a previously unrecognized autophagy-independent inhibitory function of Becn1 in iCM reprogramming.
Collapse
Affiliation(s)
- Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Peisen Huang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David Near
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yangxi Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tiffany Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chaoying Yin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael Bressan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
Bhattacharyya S, Pucadyil TJ. Cellular functions and intrinsic attributes of the ATP-binding Eps15 homology domain-containing proteins. Protein Sci 2020; 29:1321-1330. [PMID: 32223019 DOI: 10.1002/pro.3860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 01/14/2023]
Abstract
Several cellular processes rely on a cohort of dedicated proteins that manage tubulation, fission, and fusion of membranes. A notably large number of them belong to the dynamin superfamily of proteins. Among them is the evolutionarily conserved group of ATP-binding Eps15-homology domain-containing proteins (EHDs). In the two decades since their discovery, EHDs have been linked to a range of cellular processes that require remodeling or maintenance of specific membrane shapes such as during endocytic recycling, caveolar biogenesis, ciliogenesis, formation of T-tubules in skeletal muscles, and membrane resealing after rupture. Recent work has shed light on their structure and the unique attributes they possess in linking ATP hydrolysis to membrane remodeling. This review summarizes some of these recent developments and reconciles intrinsic protein functions to their cellular roles.
Collapse
Affiliation(s)
- Soumya Bhattacharyya
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Thomas J Pucadyil
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| |
Collapse
|
10
|
Tom EC, Mushtaq I, Mohapatra BC, Luan H, Bhat AM, Zutshi N, Chakraborty S, Islam N, Arya P, Bielecki TA, Iseka FM, Bhattacharyya S, Cypher LR, Goetz BT, Negi SK, Storck MD, Rana S, Barnekow A, Singh PK, Ying G, Guda C, Natarajan A, Band V, Band H. EHD1 and RUSC2 Control Basal Epidermal Growth Factor Receptor Cell Surface Expression and Recycling. Mol Cell Biol 2020; 40:e00434-19. [PMID: 31932478 PMCID: PMC7076251 DOI: 10.1128/mcb.00434-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/26/2019] [Accepted: 12/26/2019] [Indexed: 01/25/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is a prototype receptor tyrosine kinase and an oncoprotein in many solid tumors. Cell surface display of EGFR is essential for cellular responses to its ligands. While postactivation endocytic trafficking of EGFR has been well elucidated, little is known about mechanisms of basal/preactivation surface display of EGFR. Here, we identify a novel role of the endocytic regulator EHD1 and a potential EHD1 partner, RUSC2, in cell surface display of EGFR. EHD1 and RUSC2 colocalize with EGFR in vesicular/tubular structures and at the Golgi compartment. Inducible EHD1 knockdown reduced the cell surface EGFR expression with accumulation at the Golgi compartment, a phenotype rescued by exogenous EHD1. RUSC2 knockdown phenocopied the EHD1 depletion effects. EHD1 or RUSC2 depletion impaired the EGF-induced cell proliferation, demonstrating that the novel, EHD1- and RUSC2-dependent transport of unstimulated EGFR from the Golgi compartment to the cell surface that we describe is functionally important, with implications for physiologic and oncogenic roles of EGFR and targeted cancer therapies.
Collapse
Affiliation(s)
- Eric C Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bhopal C Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Aaqib M Bhat
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Neha Zutshi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sukanya Chakraborty
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Namista Islam
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Priyanka Arya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fany M Iseka
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Luke R Cypher
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Simarjeet K Negi
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Angelika Barnekow
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Guoguang Ying
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
11
|
Martins-Marques T, Catarino S, Gonçalves A, Miranda-Silva D, Gonçalves L, Antunes P, Coutinho G, Leite Moreira A, Falcão Pires I, Girão H. EHD1 Modulates Cx43 Gap Junction Remodeling Associated With Cardiac Diseases. Circ Res 2020; 126:e97-e113. [PMID: 32138615 DOI: 10.1161/circresaha.119.316502] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Efficient communication between heart cells is vital to ensure the anisotropic propagation of electrical impulses, a function mainly accomplished by gap junctions (GJ) composed of Cx43 (connexin 43). Although the molecular mechanisms remain unclear, altered distribution and function of gap junctions have been associated with acute myocardial infarction and heart failure. OBJECTIVE A recent proteomic study from our laboratory identified EHD1 (Eps15 [endocytic adaptor epidermal growth factor receptor substrate 15] homology domain-containing protein 1) as a novel interactor of Cx43 in the heart. METHODS AND RESULTS In the present work, we demonstrate that knockdown of EHD1 impaired the internalization of Cx43, preserving gap junction-intercellular coupling in cardiomyocytes. Interaction of Cx43 with EHD1 was mediated by Eps15 and promoted by phosphorylation and ubiquitination of Cx43. Overexpression of wild-type EHD1 accelerated internalization of Cx43 and exacerbated ischemia-induced lateralization of Cx43 in isolated adult cardiomyocytes. In addition, we show that EHDs associate with Cx43 in human and murine failing hearts. CONCLUSIONS Overall, we identified EHDs as novel regulators of endocytic trafficking of Cx43, participating in the pathological remodeling of gap junctions, paving the way to innovative therapeutic strategies aiming at preserving intercellular communication in the heart.
Collapse
Affiliation(s)
- Tania Martins-Marques
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Steve Catarino
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Alexandre Gonçalves
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Daniela Miranda-Silva
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Lino Gonçalves
- Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Pedro Antunes
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.).,Cardiothoracic Surgery (P.A., G.C.), Coimbra Hospital and University Centre, Portugal
| | - Gonçalo Coutinho
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.).,Cardiothoracic Surgery (P.A., G.C.), Coimbra Hospital and University Centre, Portugal
| | - Adelino Leite Moreira
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Inês Falcão Pires
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Henrique Girão
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| |
Collapse
|
12
|
Kitmitto A, Baudoin F, Cartwright EJ. Cardiomyocyte damage control in heart failure and the role of the sarcolemma. J Muscle Res Cell Motil 2019; 40:319-333. [PMID: 31520263 PMCID: PMC6831538 DOI: 10.1007/s10974-019-09539-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 01/07/2023]
Abstract
The cardiomyocyte plasma membrane, termed the sarcolemma, is fundamental for regulating a myriad of cellular processes. For example, the structural integrity of the cardiomyocyte sarcolemma is essential for mediating cardiac contraction by forming microdomains such as the t-tubular network, caveolae and the intercalated disc. Significantly, remodelling of these sarcolemma microdomains is a key feature in the development and progression of heart failure (HF). However, despite extensive characterisation of the associated molecular and ultrastructural events there is a lack of clarity surrounding the mechanisms driving adverse morphological rearrangements. The sarcolemma also provides protection, and is the cell's first line of defence, against external stresses such as oxygen and nutrient deprivation, inflammation and oxidative stress with a loss of sarcolemma viability shown to be a key step in cell death via necrosis. Significantly, cumulative cell death is also a feature of HF, and is linked to disease progression and loss of cardiac function. Herein, we will review the link between structural and molecular remodelling of the sarcolemma associated with the progression of HF, specifically considering the evidence for: (i) Whether intrinsic, evolutionary conserved, plasma membrane injury-repair mechanisms are in operation in the heart, and (ii) if deficits in key 'wound-healing' proteins (annexins, dysferlin, EHD2 and MG53) may play a yet to be fully appreciated role in triggering sarcolemma microdomain remodelling and/or necrosis. Cardiomyocytes are terminally differentiated with very limited regenerative capability and therefore preserving cell viability and cardiac function is crucially important. This review presents a novel perspective on sarcolemma remodelling by considering whether targeting proteins that regulate sarcolemma injury-repair may hold promise for developing new strategies to attenuate HF progression.
Collapse
Affiliation(s)
- Ashraf Kitmitto
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK.
| | - Florence Baudoin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| |
Collapse
|
13
|
Ponsuksili S, Trakooljul N, Basavaraj S, Hadlich F, Murani E, Wimmers K. Epigenome-wide skeletal muscle DNA methylation profiles at the background of distinct metabolic types and ryanodine receptor variation in pigs. BMC Genomics 2019; 20:492. [PMID: 31195974 PMCID: PMC6567458 DOI: 10.1186/s12864-019-5880-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Epigenetic variation may result from selection for complex traits related to metabolic processes or appear in the course of adaptation to mediate responses to exogenous stressors. Moreover epigenetic marks, in particular the DNA methylation state, of specific loci are driven by genetic variation. In this sense, polymorphism with major gene effects on metabolic and cell signaling processes, like the variation of the ryanodine receptors in skeletal muscle, may affect DNA methylation. METHODS DNA-Methylation profiles were generated applying Reduced Representation Bisulfite Sequencing (RRBS) on 17 Musculus longissimus dorsi samples. We examined DNA methylation in skeletal muscle of pig breeds differing in metabolic type, Duroc and Pietrain. We also included F2 crosses of these breeds to get a first clue to DNA methylation sites that may contribute to breed differences. Moreover, we compared DNA methylation in muscle tissue of Pietrain pigs differing in genotypes at the gene encoding the Ca2+ release channel (RYR1) that largely affects muscle physiology. RESULTS More than 2000 differently methylated sites were found between breeds including changes in methylation profiles of METRNL, IDH3B, COMMD6, and SLC22A18, genes involved in lipid metabolism. Depending on RYR1 genotype there were 1060 differently methylated sites including some functionally related genes, such as CABP2 and EHD, which play a role in buffering free cytosolic Ca2+ or interact with the Na+/Ca2+ exchanger. CONCLUSIONS The change in the level of methylation between the breeds is probably the result of the long-term selection process for quantitative traits involving an infinite number of genes, or it may be the result of a major gene mutation that plays an important role in muscle metabolism and triggers extensive compensatory processes.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany
| | - Sajjanar Basavaraj
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany
| | - Frieder Hadlich
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany
| | - Eduard Murani
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Rostock, Germany. .,Faculty of Agricultural and Environmental Sciences, University Rostock, 18059, Rostock, Germany.
| |
Collapse
|
14
|
Novel Mechanistic Roles for Ankyrin-G in Cardiac Remodeling and Heart Failure. JACC Basic Transl Sci 2018; 3:675-689. [PMID: 30456339 PMCID: PMC6234521 DOI: 10.1016/j.jacbts.2018.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/05/2018] [Accepted: 07/31/2018] [Indexed: 12/25/2022]
Abstract
The pathogenesis of human heart failure is complex, and the creation of new therapeutic strategies for human heart failure is critical. Identifying the molecular pathways underlying heart failure is important to define potential new therapeutic targets. Ankyrin polypeptides serve to target and stabilize membrane proteins in cardiomyocytes. Ankyrin-G levels are altered in humans and mice with heart failure, and mice lacking ankyrin-G in cardiomyocytes develop cardiomyopathy and systolic dysfunction. Mechanistically, ankyrin-G is necessary for the expression and localization of critical myocyte proteins essential for regulating cardiac structural and electrical activity.
Ankyrin polypeptides are intracellular proteins responsible for targeting cardiac membrane proteins. Here, the authors demonstrate that ankyrin-G plays an unexpected role in normal compensatory physiological remodeling in response to myocardial stress and aging; the authors implicate disruption of ankyrin-G in human heart failure. Mechanistically, the authors illustrate that ankyrin-G serves as a key nodal protein required for cardiac myofilament integration with the intercalated disc. Their data define novel in vivo mechanistic roles for ankyrin-G, implicate ankyrin-G as necessary for compensatory cardiac physiological remodeling under stress, and implicate disruption of ankyrin-G in the development and progression of human heart failure.
Collapse
Key Words
- AnkG, ankyrin-G
- DSP, desmoplakin
- ECG, electrocardiogram
- HF, heart failure
- LV, left ventricular
- Nav1.5
- PBS, phosphate-buffered saline
- PKP2, plakophilin-2
- TAC, transverse aortic constriction
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick-end labeling
- WT, wild-type
- ankyrin
- arrhythmia
- cKO, cardiomyocyte-specific knockout
- cytoskeleton
- heart failure
- ion channel
Collapse
|
15
|
Dun W, Danilo P, Mohler PJ, Boyden PA. Microtubular remodeling and decreased expression of Nav1.5 with enhanced EHD4 in cells from the infarcted heart. Life Sci 2018. [DOI: 10.1016/j.lfs.2018.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
16
|
A personalized, multiomics approach identifies genes involved in cardiac hypertrophy and heart failure. NPJ Syst Biol Appl 2018; 4:12. [PMID: 29507758 PMCID: PMC5825397 DOI: 10.1038/s41540-018-0046-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 12/14/2017] [Accepted: 01/12/2018] [Indexed: 11/08/2022] Open
Abstract
A traditional approach to investigate the genetic basis of complex diseases is to identify genes with a global change in expression between diseased and healthy individuals. However, population heterogeneity may undermine the effort to uncover genes with significant but individual contribution to the spectrum of disease phenotypes within a population. Here we investigate individual changes of gene expression when inducing hypertrophy and heart failure in 100 + strains of genetically distinct mice from the Hybrid Mouse Diversity Panel (HMDP). We find that genes whose expression fold-change correlates in a statistically significant way with the severity of the disease are either up or down-regulated across strains, and therefore missed by a traditional population-wide analysis of differential gene expression. Furthermore, those "fold-change" genes are enriched in human cardiac disease genes and form a dense co-regulated module strongly interacting with the cardiac hypertrophic signaling network in the human interactome. We validate our approach by showing that the knockdown of Hes1, predicted as a strong candidate, induces a dramatic reduction of hypertrophy by 80-90% in neonatal rat ventricular myocytes. Our results demonstrate that individualized approaches are crucial to identify genes underlying complex diseases as well as to develop personalized therapies.
Collapse
|
17
|
Rask-Andersen M, Martinsson D, Ahsan M, Enroth S, Ek WE, Gyllensten U, Johansson Å. Epigenome-wide association study reveals differential DNA methylation in individuals with a history of myocardial infarction. Hum Mol Genet 2018; 25:4739-4748. [PMID: 28172975 DOI: 10.1093/hmg/ddw302] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide and represent a substantial economic burden on public health care systems. Epigenetic markers have potential as diagnostic markers before clinical symptoms have emerged, and as prognostic markers to inform the choice of clinical intervention. In this study, we performed an epigenome-wide association study (EWAS) for CVDs, to identify disease-specific alterations in DNA methylation. CpG methylation in blood samples from the northern Sweden population health study (NSPHS) (n = 729) was assayed on the Illumina Infinium HumanMethylation450 BeadChip. Individuals with a history of a CVD were identified in the cohort. It included individuals with hypertension (N = 147), myocardial infarction (MI) (N = 48), stroke (N = 27), thrombosis (N = 22) and cardiac arrhythmia (N = 5). Differential DNA methylation was observed at 211 CpG-sites in individuals with a history of MI (q <0.05). These sites represent 196 genes, of which 42 have been described in the scientific literature to be related to cardiac function, cardiovascular disease, cardiogenesis and recovery after ischemic injury. We have shown that individuals with a history of MI have a deviating pattern of DNA methylation at many genomic loci of which a large fraction has previously been linked to CVD. Our results highlight genes that might be important in the pathogenesis of MI or in recovery. In addition, the sites pointed out in this study can serve as candidates for further evaluation as potential biomarkers for MI.
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - David Martinsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muhammad Ahsan
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Enroth
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Weronica E Ek
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Nair NU, Das A, Amit U, Robinson W, Park SG, Basu M, Lugo A, Leor J, Ruppin E, Hannenhalli S. Putative functional genes in idiopathic dilated cardiomyopathy. Sci Rep 2018; 8:66. [PMID: 29311597 PMCID: PMC5758757 DOI: 10.1038/s41598-017-18524-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Idiopathic dilated cardiomyopathy (DCM) is a complex disorder with a genetic and an environmental component involving multiple genes, many of which are yet to be discovered. We integrate genetic, epigenetic, transcriptomic, phenotypic, and evolutionary features into a method - Hridaya, to infer putative functional genes underlying DCM in a genome-wide fashion, using 213 human heart genomes and transcriptomes. Many genes identified by Hridaya are experimentally shown to cause cardiac complications. We validate the top predicted genes, via five different genome-wide analyses: First, the predicted genes are associated with cardiovascular functions. Second, their knockdowns in mice induce cardiac abnormalities. Third, their inhibition by drugs cause cardiac side effects in human. Fourth, they tend to have differential exon usage between DCM and normal samples. Fifth, analyzing 213 individual genotypes, we show that regulatory polymorphisms of the predicted genes are associated with elevated risk of cardiomyopathy. The stratification of DCM patients based on cardiac expression of the functional genes reveals two subgroups differing in key cardiac phenotypes. Integrating predicted functional genes with cardiomyocyte drug treatment experiments reveals novel potential drug targets. We provide a list of investigational drugs that target the newly identified functional genes that may lead to cardiac side effects.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA.
| | - Avinash Das
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
| | - Uri Amit
- The Neufeld Cardiac Research Institute, Tel Aviv University, Tel Aviv-Yafo, Israel
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Ramat Gan, Israel
- The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Radiation Oncology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Welles Robinson
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
| | - Seung Gu Park
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
| | - Mahashweta Basu
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
| | - Alex Lugo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
| | - Jonathan Leor
- The Neufeld Cardiac Research Institute, Tel Aviv University, Tel Aviv-Yafo, Israel
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Eytan Ruppin
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, 20742, USA.
| |
Collapse
|
19
|
Yang HQ, Jana K, Rindler MJ, Coetzee WA. The trafficking protein, EHD2, positively regulates cardiac sarcolemmal K ATP channel surface expression: role in cardioprotection. FASEB J 2018; 32:1613-1625. [PMID: 29133341 DOI: 10.1096/fj.201700027r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ATP-sensitive K+ (KATP) channels uniquely link cellular energy metabolism to membrane excitability and are expressed in diverse cell types that range from the endocrine pancreas to neurons and smooth, skeletal, and cardiac muscle. A decrease in the surface expression of KATP channels has been linked to various disorders, including dysregulated insulin secretion, abnormal blood pressure, and impaired resistance to cardiac injury. In contrast, up-regulation of KATP channel surface expression may be protective, for example, by mediating the beneficial effect of ischemic preconditioning. Molecular mechanisms that regulate KATP channel trafficking are poorly understood. Here, we used cellular assays with immunofluorescence, surface biotinylation, and patch clamping to demonstrate that Eps15 homology domain-containing protein 2 (EHD2) is a novel positive regulator of KATP channel trafficking to increase surface KATP channel density. EHD2 had no effect on cardiac Na+ channels (Nav1.5). The effect is specific to EHD2 as other members of the EHD family-EHD1, EHD3, and EHD4-had no effect on KATP channel surface expression. EHD2 did not directly affect KATP channel properties as unitary conductance and ATP sensitivity were unchanged. Instead, we observed that the mechanism by which EHD2 increases surface expression is by stabilizing KATP channel-containing caveolar structures, which results in a reduced rate of endocytosis. EHD2 also regulated KATP channel trafficking in isolated cardiomyocytes, which validated the physiologic relevance of these observations. Pathophysiologically, EHD2 may be cardioprotective as a dominant-negative EHD2 mutant sensitized cardiomyocytes to ischemic damage. Our findings highlight EHD2 as a potential pharmacologic target in the treatment of diseases with KATP channel trafficking defects.-Yang, H. Q., Jana, K., Rindler, M. J., Coetzee, W. A. The trafficking protein, EHD2, positively regulates cardiac sarcolemmal KATP channel surface expression: role in cardioprotection.
Collapse
Affiliation(s)
- Hua Qian Yang
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Kundan Jana
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Michael J Rindler
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - William A Coetzee
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA.,Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York, USA.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Iseka FM, Goetz BT, Mushtaq I, An W, Cypher LR, Bielecki TA, Tom EC, Arya P, Bhattacharyya S, Storck MD, Semerad CL, Talmadge JE, Mosley RL, Band V, Band H. Role of the EHD Family of Endocytic Recycling Regulators for TCR Recycling and T Cell Function. THE JOURNAL OF IMMUNOLOGY 2017; 200:483-499. [PMID: 29212907 DOI: 10.4049/jimmunol.1601793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/01/2017] [Indexed: 12/31/2022]
Abstract
T cells use the endocytic pathway for key cell biological functions, including receptor turnover and maintenance of the immunological synapse. Some of the established players include the Rab GTPases, the SNARE complex proteins, and others, which function together with EPS-15 homology domain-containing (EHD) proteins in non-T cell systems. To date, the role of the EHD protein family in T cell function remains unexplored. We generated conditional EHD1/3/4 knockout mice using CD4-Cre and crossed these with mice bearing a myelin oligodendrocyte glycoprotein-specific TCR transgene. We found that CD4+ T cells from these mice exhibited reduced Ag-driven proliferation and IL-2 secretion in vitro. In vivo, these mice exhibited reduced severity of experimental autoimmune encephalomyelitis. Further analyses showed that recycling of the TCR-CD3 complex was impaired, leading to increased lysosomal targeting and reduced surface levels on CD4+ T cells of EHD1/3/4 knockout mice. Our studies reveal a novel role of the EHD family of endocytic recycling regulatory proteins in TCR-mediated T cell functions.
Collapse
Affiliation(s)
- Fany M Iseka
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Luke R Cypher
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Eric C Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Priyanka Arya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Craig L Semerad
- Flow Cytometry Research Facility, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - R Lee Mosley
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198; .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
21
|
Swayne LA, Murphy NP, Asuri S, Chen L, Xu X, McIntosh S, Wang C, Lancione PJ, Roberts JD, Kerr C, Sanatani S, Sherwin E, Kline CF, Zhang M, Mohler PJ, Arbour LT. Novel Variant in the ANK2 Membrane-Binding Domain Is Associated With Ankyrin-B Syndrome and Structural Heart Disease in a First Nations Population With a High Rate of Long QT Syndrome. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.116.001537. [PMID: 28196901 DOI: 10.1161/circgenetics.116.001537] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/21/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Long QT syndrome confers susceptibility to ventricular arrhythmia, predisposing to syncope, seizures, and sudden death. While rare globally, long QT syndrome is ≈15× more common in First Nations of Northern British Columbia largely because of a known mutation in KCNQ1. However, 2 large multigenerational families were affected, but negative for the known mutation. METHODS AND RESULTS Long QT syndrome panel testing was carried out in the index case of each family, and clinical information was collected. Cascade genotyping was performed. Biochemical and myocyte-based assays were performed to evaluate the identified gene variant for loss-of-function activity. Index cases in these 2 families harbored a novel ANK2 c.1937C>T variant (p.S646F). An additional 16 carriers were identified, including 2 with structural heart disease: one with cardiomyopathy resulting in sudden death and the other with congenital heart disease. For all carriers of this variant, the average QTc was 475 ms (±40). Although ankyrin-B p.S646F is appropriately folded and expressed in bacteria, the mutant polypeptide displays reduced expression in cultured H9c2 cells and aberrant localization in primary cardiomyocytes. Furthermore, myocytes expressing ankyrin-B p.S646F lack normal membrane targeting of the ankyrin-binding partner, the Na/Ca exchanger. Thus, ankyrin-B p.S646F is a loss-of-function variant. CONCLUSIONS We identify the first disease-causing ANK2 variant localized to the membrane-binding domain resulting in reduced ankyrin-B expression and abnormal localization. Further study is warranted on the potential association of this variant with structural heart disease given the role of ANK2 in targeting and stabilization of key structural and signaling molecules in cardiac cells.
Collapse
Affiliation(s)
- Leigh Anne Swayne
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Nathaniel P Murphy
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Sirisha Asuri
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Lena Chen
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Xiaoxue Xu
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Sarah McIntosh
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Chao Wang
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Peter J Lancione
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Jason D Roberts
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Charles Kerr
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Shubhayan Sanatani
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Elizabeth Sherwin
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Crystal F Kline
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Mingjie Zhang
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Peter J Mohler
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.)
| | - Laura T Arbour
- From the Division of Medical Sciences, University of Victoria, BC, Canada (L.A.S., L.C., X.X., L.T.A.); University of British Columbia Island Medical Program, Victoria, BC, Canada (L.A.S., L.T.A.); Department of Medical Genetics (S.A., S.M., L.T.A.), Division of Cardiology (C.K.), and Division of Cardiology, Department of Pediatrics, BC Children's Hospital (S.S., E.S.), University of British Columbia, Vancouver, BC, Canada; Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute (N.P.M., P.J.L., C.F.K., P.J.M.) and Department of Physiology and Cell Biology (N.P.M., P.J.L., C.F.K., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China (C.W., M.Z.); and Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada (J.D.R.).
| |
Collapse
|
22
|
Semmler J, Kormann J, Srinivasan SP, Köster A, Sälzer D, Reppel M, Hescheler J, Plomann M, Nguemo F. Pacsin 2 is required for the maintenance of a normal cardiac function in the developing mouse heart. Pharmacol Res 2017; 128:200-210. [PMID: 29107716 DOI: 10.1016/j.phrs.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/26/2017] [Accepted: 10/15/2017] [Indexed: 11/27/2022]
Abstract
The Pacsin proteins (Pacsin 1, 2 and 3) play an important role in intracellular trafficking and thereby signal transduction in many cells types. This study was designed to examine the role of Pacsin 2 in cardiac development and function. We investigated the development and electrophysiological properties of Pacsin 2 knockout (P2KO) hearts and single cardiomyocytes isolated from 11.5 and 15.5days old fetal mice. Immunofluorescence experiments confirmed the lack of Pacsin 2 protein expression in P2KO cardiac myocytes in comparison to wildtype (WT). Western blotting demonstrates low expression levels of connexin 43 and T-box 3 proteins in P2KO compared to wildtype (WT). Electrophysiology measurements including online Multi-Electrode Array (MEA) based field potential (FP) recordings on isolated whole heart of P2KO mice showed a prolonged AV-conduction time. Patch clamp measurements of P2KO cardiomyocytes revealed differences in action potential (AP) parameters and decreased pacemaker funny channel (If), as well as L-type Ca2+ channel (ICaL), and sodium channel (INa). These findings demonstrate that Pacsin 2 is necessary for cardiac development and function in mouse embryos, which will enhance our knowledge to better understand the genesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Judith Semmler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Jan Kormann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Annette Köster
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Daniel Sälzer
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany; Department of Cardiology, University of Lübeck, Lübeck, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Markus Plomann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
23
|
Rahman SS, Moffitt AEJ, Trease AJ, Foster KW, Storck MD, Band H, Boesen EI. EHD4 is a novel regulator of urinary water homeostasis. FASEB J 2017; 31:5217-5233. [PMID: 28778975 DOI: 10.1096/fj.201601182rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 07/25/2017] [Indexed: 01/08/2023]
Abstract
The Eps15-homology domain-containing (EHD) protein family comprises 4 members that regulate endocytic recycling. Although the kidney expresses all 4 EHD proteins, their physiologic roles are largely unknown. This study focused on EHD4, which we found to be expressed differentially across nephron segments with the highest expression in the inner medullary collecting duct. Under baseline conditions, Ehd4-/- [EHD4-knockout (KO)] mice on a C57Bl/6 background excreted a higher volume of more dilute urine than control C57Bl/6 wild-type (WT) mice while maintaining a similar plasma osmolality. Urine excretion after an acute intraperitoneal water load was significantly increased in EHD4-KO mice compared to WT mice, and although EHD4-KO mice concentrated their urine during 24-h water restriction, urinary osmolality remained significantly lower than in WT mice, suggesting that EHD4 plays a role in renal water handling. Total aquaporin 2 (AQP2) and phospho-S256-AQP2 (pAQP2) protein expression in the inner medulla was similar in the two groups in baseline conditions. However, localization of both AQP2 and pAQP2 in the renal inner medullary principal cells appeared more dispersed, and the intensity of apical membrane staining for AQP2 was reduced significantly (by ∼20%) in EHD4-KO mice compared to WT mice in baseline conditions, suggesting an important role of EHD4 in trafficking of AQP2. Together, these data indicate that EHD4 play important roles in the regulation of water homeostasis.-Rahman, S. S., Moffitt, A. E. J., Trease, A. J., Foster, K. W., Storck, M. D., Band, H., Boesen, E. I. EHD4 is a novel regulator of urinary water homeostasis.
Collapse
Affiliation(s)
- Shamma S Rahman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Alexandra E J Moffitt
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew D Storck
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Hamid Band
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA; .,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA; and.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| |
Collapse
|
24
|
Abstract
Over the past decade, ankyrin-B has been identified as a prominent player in cardiac physiology. Ankyrin-B has a multitude of functions, with roles in expression, localization, and regulation of proteins critical for cardiac excitability, cytoskeletal integrity, and signaling. Furthermore, human ANK2 variants that result in ankyrin-B loss of function are associated with "ankyrin-B syndrome," a complex cardiac phenotype that may include bradycardia and heart rate variability, conduction block, atrial fibrillation, QT interval prolongation, and potentially fatal catecholaminergic polymorphic ventricular tachycardia. However, our understanding of the molecular mechanisms underlying ankyrin-B function at baseline and in disease is still not fully developed owing to the complexity of ankyrin-B gene regulation, number of ankyrin-B-associated molecules, multiple roles of ankyrin-B in the heart and other organs that modulate cardiac function, and a host of unexpected clinical phenotypes. In this review, we summarize known roles of ankyrin-B in the heart and the impact of ankyrin-B dysfunction in animal models and in human disease as well as highlight important new findings illustrating the complexity of ankyrin-B signaling.
Collapse
Affiliation(s)
- Sara N Koenig
- Dorothy M. Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio.
| | - Peter J Mohler
- Dorothy M. Davis Heart & Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
25
|
Pinali C, Malik N, Davenport JB, Allan LJ, Murfitt L, Iqbal MM, Boyett MR, Wright EJ, Walker R, Zhang Y, Dobryznski H, Holt CM, Kitmitto A. Post-Myocardial Infarction T-tubules Form Enlarged Branched Structures With Dysregulation of Junctophilin-2 and Bridging Integrator 1 (BIN-1). J Am Heart Assoc 2017; 6:e004834. [PMID: 28473402 PMCID: PMC5524063 DOI: 10.1161/jaha.116.004834] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/26/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart failure is a common secondary complication following a myocardial infarction (MI), characterized by impaired cardiac contraction and t-tubule (t-t) loss. However, post-MI nano-scale morphological changes to the remaining t-ts are poorly understood. METHOD AND RESULTS We utilized a porcine model of MI, using a nonlethal microembolization method to generate controlled microinfarcts. Using serial block face scanning electron microscopy, we report that post-MI, after mild left-ventricular dysfunction has developed, t-ts are not only lost in the peri-infarct region, but also the remnant t-ts form enlarged, highly branched disordered structures, containing a dense intricate inner membrane. Biochemical and proteomics analyses showed that the calcium release channel, ryanodine receptor 2 (RyR2), abundance is unchanged, but junctophilin-2 (JP2), important for maintaining t-t trajectory, is depressed (-0.5×) in keeping with the t-ts being disorganized. However, immunolabeling shows that populations of RyR2 and JP2 remain associated with the remodeled t-ts. The bridging integrator 1 protein (BIN-1), a regulator of tubulogensis, is upregulated (+5.4×), consistent with an overdeveloped internal membrane system, a feature not present in control t-ts. Importantly, we have determined that t-ts, in the remote region, are narrowed and also contain dense membrane folds (BIN-1 is up-regulated +3.4×), whereas the t-ts have a radial organization comparable to control JP2 is upregulated +1.7×. CONCLUSIONS This study reveals previously unidentified remodeling of the t-t nano-architecture in the post-MI heart that extends to the remote region. Our findings highlight that targeting JP2 may be beneficial for preserving the orientation of the t-ts, attenuating the development of hypocontractility post-MI.
Collapse
Affiliation(s)
- Christian Pinali
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Nadim Malik
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - J Bernard Davenport
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Laurence J Allan
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Lucy Murfitt
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Mohammad M Iqbal
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Mark R Boyett
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Elizabeth J Wright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Rachel Walker
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Yu Zhang
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Halina Dobryznski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Cathy M Holt
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| |
Collapse
|
26
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Bhattacharyya S, Rainey MA, Arya P, Mohapatra BC, Mushtaq I, Dutta S, George M, Storck MD, McComb RD, Muirhead D, Todd GL, Gould K, Datta K, Gelineau-van Waes J, Band V, Band H. Endocytic recycling protein EHD1 regulates primary cilia morphogenesis and SHH signaling during neural tube development. Sci Rep 2016; 6:20727. [PMID: 26884322 PMCID: PMC4756679 DOI: 10.1038/srep20727] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
Members of the four-member C-terminal EPS15-Homology Domain-containing (EHD) protein family play crucial roles in endocytic recycling of cell surface receptors from endosomes to the plasma membrane. In this study, we show that Ehd1 gene knockout in mice on a predominantly B6 background is embryonic lethal. Ehd1-null embryos die at mid-gestation with a failure to complete key developmental processes including neural tube closure, axial turning and patterning of the neural tube. We found that Ehd1-null embryos display short and stubby cilia on the developing neuroepithelium at embryonic day 9.5 (E9.5). Loss of EHD1 also deregulates the ciliary SHH signaling with Ehd1-null embryos displaying features indicative of increased SHH signaling, including a significant downregulation in the formation of the GLI3 repressor and increase in the ventral neuronal markers specified by SHH. Using Ehd1-null MEFS we found that EHD1 protein co-localizes with the SHH receptor Smoothened in the primary cilia upon ligand stimulation. Under the same conditions, EHD1 was shown to co-traffic with Smoothened into the developing primary cilia and we identify EHD1 as a direct binding partner of Smoothened. Overall, our studies identify the endocytic recycling regulator EHD1 as a novel regulator of the primary cilium-associated trafficking of Smoothened and Hedgehog signaling.
Collapse
Affiliation(s)
- Sohinee Bhattacharyya
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark A Rainey
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Priyanka Arya
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Samikshan Dutta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney D McComb
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Muirhead
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gordon L Todd
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Karen Gould
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaustubh Datta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Vimla Band
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hamid Band
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
28
|
An Adaptable Spectrin/Ankyrin-Based Mechanism for Long-Range Organization of Plasma Membranes in Vertebrate Tissues. CURRENT TOPICS IN MEMBRANES 2015; 77:143-84. [PMID: 26781832 DOI: 10.1016/bs.ctm.2015.10.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ankyrins are membrane-associated proteins that together with their spectrin partners are responsible for micron-scale organization of vertebrate plasma membranes, including those of erythrocytes, excitable membranes of neurons and heart, lateral membrane domains of columnar epithelial cells, and striated muscle. Ankyrins coordinate functionally related membrane transporters and cell adhesion proteins (15 protein families identified so far) within plasma membrane compartments through independently evolved interactions of intrinsically disordered sequences with a highly conserved peptide-binding groove formed by the ANK repeat solenoid. Ankyrins are coupled to spectrins, which are elongated organelle-sized proteins that form mechanically resilient arrays through cross-linking by specialized actin filaments. In addition to protein interactions, cellular targeting and assembly of spectrin/ankyrin domains also critically depend on palmitoylation of ankyrin-G by aspartate-histidine-histidine-cysteine 5/8 palmitoyltransferases, as well as interaction of beta-2 spectrin with phosphoinositide lipids. These lipid-dependent spectrin/ankyrin domains are not static but are locally dynamic and determine membrane identity through opposing endocytosis of bulk lipids as well as specific proteins. A partnership between spectrin, ankyrin, and cell adhesion molecules first emerged in bilaterians over 500 million years ago. Ankyrin and spectrin may have been recruited to plasma membranes from more ancient roles in organelle transport. The basic bilaterian spectrin-ankyrin toolkit markedly expanded in vertebrates through gene duplications combined with variation in unstructured intramolecular regulatory sequences as well as independent evolution of ankyrin-binding activity by ion transporters involved in action potentials and calcium homeostasis. In addition, giant vertebrate ankyrins with specialized roles in axons acquired new coding sequences by exon shuffling. We speculate that early axon initial segments and epithelial lateral membranes initially were based on spectrin-ankyrin-cell adhesion molecule assemblies and subsequently served as "incubators," where ion transporters independently acquired ankyrin-binding activity through positive selection.
Collapse
|
29
|
Arya P, Rainey MA, Bhattacharyya S, Mohapatra BC, George M, Kuracha MR, Storck MD, Band V, Govindarajan V, Band H. The endocytic recycling regulatory protein EHD1 Is required for ocular lens development. Dev Biol 2015; 408:41-55. [PMID: 26455409 DOI: 10.1016/j.ydbio.2015.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 09/01/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022]
Abstract
The C-terminal Eps15 homology domain-containing (EHD) proteins play a key role in endocytic recycling, a fundamental cellular process that ensures the return of endocytosed membrane components and receptors back to the cell surface. To define the in vivo biological functions of EHD1, we have generated Ehd1 knockout mice and previously reported a requirement of EHD1 for spermatogenesis. Here, we show that approximately 56% of the Ehd1-null mice displayed gross ocular abnormalities, including anophthalmia, aphakia, microphthalmia and congenital cataracts. Histological characterization of ocular abnormalities showed pleiotropic defects that include a smaller or absent lens, persistence of lens stalk and hyaloid vasculature, and deformed optic cups. To test whether these profound ocular defects resulted from the loss of EHD1 in the lens or in non-lenticular tissues, we deleted the Ehd1 gene selectively in the presumptive lens ectoderm using Le-Cre. Conditional Ehd1 deletion in the lens resulted in developmental defects that included thin epithelial layers, small lenses and absence of corneal endothelium. Ehd1 deletion in the lens also resulted in reduced lens epithelial proliferation, survival and expression of junctional proteins E-cadherin and ZO-1. Finally, Le-Cre-mediated deletion of Ehd1 in the lens led to defects in corneal endothelial differentiation. Taken together, these data reveal a unique role for EHD1 in early lens development and suggest a previously unknown link between the endocytic recycling pathway and regulation of key developmental processes including proliferation, differentiation and morphogenesis.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Mark A Rainey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center Omaha, NE 68198-5900, USA.
| | - Bhopal C Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center Omaha, NE 68198-5870, USA.
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Murali R Kuracha
- Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Vimla Band
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA.
| | - Venkatesh Govindarajan
- Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Hamid Band
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center Omaha, NE 68198-5900, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA.
| |
Collapse
|
30
|
Demonbreun AR, Swanson KE, Rossi AE, Deveaux HK, Earley JU, Allen MV, Arya P, Bhattacharyya S, Band H, Pytel P, McNally EM. Eps 15 Homology Domain (EHD)-1 Remodels Transverse Tubules in Skeletal Muscle. PLoS One 2015; 10:e0136679. [PMID: 26325203 PMCID: PMC4556691 DOI: 10.1371/journal.pone.0136679] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/06/2015] [Indexed: 11/19/2022] Open
Abstract
We previously showed that Eps15 homology domain-containing 1 (EHD1) interacts with ferlin proteins to regulate endocytic recycling. Myoblasts from Ehd1-null mice were found to have defective recycling, myoblast fusion, and consequently smaller muscles. When expressed in C2C12 cells, an ATPase dead-EHD1 was found to interfere with BIN1/amphiphysin 2. We now extended those findings by examining Ehd1-heterozygous mice since these mice survive to maturity in normal Mendelian numbers and provide a ready source of mature muscle. We found that heterozygosity of EHD1 was sufficient to produce ectopic and excessive T-tubules, including large intracellular aggregates that contained BIN1. The disorganized T-tubule structures in Ehd1-heterozygous muscle were accompanied by marked elevation of the T-tubule-associated protein DHPR and reduction of the triad linker protein junctophilin 2, reflecting defective triads. Consistent with this, Ehd1-heterozygous muscle had reduced force production. Introduction of ATPase dead-EHD1 into mature muscle fibers was sufficient to induce ectopic T-tubule formation, seen as large BIN1 positive structures throughout the muscle. Ehd1-heterozygous mice were found to have strikingly elevated serum creatine kinase and smaller myofibers, but did not display findings of muscular dystrophy. These data indicate that EHD1 regulates the maintenance of T-tubules through its interaction with BIN1 and links T-tubules defects with elevated creatine kinase and myopathy.
Collapse
Affiliation(s)
- Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States of America
- * E-mail:
| | - Kaitlin E. Swanson
- Department of Pathology, The University of Chicago, Chicago, IL, United States of America
| | - Ann E. Rossi
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - H. Kieran Deveaux
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Judy U. Earley
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States of America
| | - Madison V. Allen
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States of America
| | - Priyanka Arya
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Sohinee Bhattacharyya
- Department of Pathology & Microbiology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Hamid Band
- Department of Pathology & Microbiology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL, United States of America
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States of America
| |
Collapse
|
31
|
Lorenzo DN, Healy JA, Hostettler J, Davis J, Yang J, Wang C, Hohmeier HE, Zhang M, Bennett V. Ankyrin-B metabolic syndrome combines age-dependent adiposity with pancreatic β cell insufficiency. J Clin Invest 2015; 125:3087-102. [PMID: 26168218 DOI: 10.1172/jci81317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Rare functional variants of ankyrin-B have been implicated in human disease, including hereditary cardiac arrhythmia and type 2 diabetes (T2D). Here, we developed murine models to evaluate the metabolic consequences of these alterations in vivo. Specifically, we generated knockin mice that express either the human ankyrin-B variant R1788W, which is present in 0.3% of North Americans of mixed European descent and is associated with T2D, or L1622I, which is present in 7.5% of African Americans. Young AnkbR1788W/R1788W mice displayed primary pancreatic β cell insufficiency that was characterized by reduced insulin secretion in response to muscarinic agonists, combined with increased peripheral glucose uptake and concomitantly increased plasma membrane localization of glucose transporter 4 (GLUT4) in skeletal muscle and adipocytes. In contrast, older AnkbR1788W/R1788W and AnkbL1622I/L1622I mice developed increased adiposity, a phenotype that was reproduced in cultured adipocytes, and insulin resistance. GLUT4 trafficking was altered in animals expressing mutant forms of ankyrin-B, and we propose that increased cell surface expression of GLUT4 in skeletal muscle and fatty tissue of AnkbR1788W/R1788W mice leads to the observed age-dependent adiposity. Together, our data suggest that ankyrin-B deficiency results in a metabolic syndrome that combines primary pancreatic β cell insufficiency with peripheral insulin resistance and is directly relevant to the nearly one million North Americans bearing the R1788W ankyrin-B variant.
Collapse
|
32
|
Sturm AC, Kline CF, Glynn P, Johnson BL, Curran J, Kilic A, Higgins RSD, Binkley PF, Janssen PML, Weiss R, Raman SV, Fowler SJ, Priori SG, Hund TJ, Carnes CA, Mohler PJ. Use of whole exome sequencing for the identification of Ito-based arrhythmia mechanism and therapy. J Am Heart Assoc 2015; 4:JAHA.114.001762. [PMID: 26015324 PMCID: PMC4599408 DOI: 10.1161/jaha.114.001762] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Identified genetic variants are insufficient to explain all cases of inherited arrhythmia. We tested whether the integration of whole exome sequencing with well-established clinical, translational, and basic science platforms could provide rapid and novel insight into human arrhythmia pathophysiology and disease treatment. METHODS AND RESULTS We report a proband with recurrent ventricular fibrillation, resistant to standard therapeutic interventions. Using whole-exome sequencing, we identified a variant in a previously unidentified exon of the dipeptidyl aminopeptidase-like protein-6 (DPP6) gene. This variant is the first identified coding mutation in DPP6 and augments cardiac repolarizing current (Ito) causing pathological changes in Ito and action potential morphology. We designed a therapeutic regimen incorporating dalfampridine to target Ito. Dalfampridine, approved for multiple sclerosis, normalized the ECG and reduced arrhythmia burden in the proband by >90-fold. This was combined with cilostazol to accelerate the heart rate to minimize the reverse-rate dependence of augmented Ito. CONCLUSIONS We describe a novel arrhythmia mechanism and therapeutic approach to ameliorate the disease. Specifically, we identify the first coding variant of DPP6 in human ventricular fibrillation. These findings illustrate the power of genetic approaches for the elucidation and treatment of disease when carefully integrated with clinical and basic/translational research teams.
Collapse
Affiliation(s)
- Amy C Sturm
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.)
| | - Crystal F Kline
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (C.F.K., J.C., P.L.J., P.J.M.)
| | - Patric Glynn
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Surgery, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (P.G., A.K., R.D.H.)
| | - Benjamin L Johnson
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.)
| | - Jerry Curran
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (C.F.K., J.C., P.L.J., P.J.M.)
| | - Ahmet Kilic
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Surgery, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (P.G., A.K., R.D.H.)
| | - Robert S D Higgins
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Surgery, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (P.G., A.K., R.D.H.)
| | - Philip F Binkley
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.)
| | - Paul M L Janssen
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (C.F.K., J.C., P.L.J., P.J.M.)
| | - Raul Weiss
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.)
| | - Subha V Raman
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.)
| | - Steven J Fowler
- Cardiovascular Genetics Program, Clinical Cardiac Electrophysiology, New York University Langone Medical Center, New York, NY (S.J.F., S.G.P.) Leon H. Charney Division of Cardiology, New York University Langone Medical Center, New York, NY (S.J.F.)
| | - Silvia G Priori
- Cardiovascular Genetics Program, Clinical Cardiac Electrophysiology, New York University Langone Medical Center, New York, NY (S.J.F., S.G.P.) Molecular Cardiology, IRCCS Fondazione Salvatore Maugeri, University of Pavia, Italy (S.G.P.) Department of Cardiology, University of Pavia, Italy (S.G.P.)
| | - Thomas J Hund
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.) Department of Biomedical Engineering, The Ohio State University, Columbus, OH (T.J.H.)
| | - Cynthia A Carnes
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) College of Pharmacy, Columbus, OH (C.A.C.)
| | - Peter J Mohler
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., C.F.K., P.G., B.L.J., J.C., A.K., R.D.H., P.F.B., P.L.J., R.W., S.V.R., T.J.H., C.A.C., P.J.M.) Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (A.C.S., P.F.B., R.W., S.V.R., T.J.H., P.J.M.) Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, The Ohio State University College of Engineering, Columbus, OH (C.F.K., J.C., P.L.J., P.J.M.)
| |
Collapse
|
33
|
Curran J, Musa H, Kline CF, Makara MA, Little SC, Higgins JD, Hund TJ, Band H, Mohler PJ. Eps15 Homology Domain-containing Protein 3 Regulates Cardiac T-type Ca2+ Channel Targeting and Function in the Atria. J Biol Chem 2015; 290:12210-21. [PMID: 25825486 DOI: 10.1074/jbc.m115.646893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Indexed: 11/06/2022] Open
Abstract
Proper trafficking of membrane-bound ion channels and transporters is requisite for normal cardiac function. Endosome-based protein trafficking of membrane-bound ion channels and transporters in the heart is poorly understood, particularly in vivo. In fact, for select cardiac cell types such as atrial myocytes, virtually nothing is known regarding endosomal transport. We previously linked the C-terminal Eps15 homology domain-containing protein 3 (EHD3) with endosome-based protein trafficking in ventricular cardiomyocytes. Here we sought to define the roles and membrane protein targets for EHD3 in atria. We identify the voltage-gated T-type Ca(2+) channels (CaV3.1, CaV3.2) as substrates for EHD3-dependent trafficking in atria. Mice selectively lacking EHD3 in heart display reduced expression and targeting of both Cav3.1 and CaV3.2 in the atria. Furthermore, functional experiments identify a significant loss of T-type-mediated Ca(2+) current in EHD3-deficient atrial myocytes. Moreover, EHD3 associates with both CaV3.1 and CaV3.2 in co-immunoprecipitation experiments. T-type Ca(2+) channel function is critical for proper electrical conduction through the atria. Consistent with these roles, EHD3-deficient mice demonstrate heart rate variability, sinus pause, and atrioventricular conduction block. In summary, our findings identify CaV3.1 and CaV3.2 as substrates for EHD3-dependent protein trafficking in heart, provide in vivo data on endosome-based trafficking pathways in atria, and implicate EHD3 as a key player in the regulation of atrial myocyte excitability and cardiac conduction.
Collapse
Affiliation(s)
- Jerry Curran
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology,
| | - Hassan Musa
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology
| | - Crystal F Kline
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology
| | - Michael A Makara
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology
| | - Sean C Little
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology
| | - John D Higgins
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology
| | - Thomas J Hund
- From the Dorothy M. Davis Heart and Lung Research Institute, Biomedical Engineering,The Ohio State University Wexner Medical Center, Columbus, Ohio 43210 and
| | - Hamid Band
- The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Peter J Mohler
- From the Dorothy M. Davis Heart and Lung Research Institute, the Departments of Physiology and Cell Biology, Medicine, and
| |
Collapse
|
34
|
Curran J, Makara MA, Mohler PJ. Endosome-based protein trafficking and Ca(2+) homeostasis in the heart. Front Physiol 2015; 6:34. [PMID: 25709583 PMCID: PMC4321435 DOI: 10.3389/fphys.2015.00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/21/2015] [Indexed: 11/13/2022] Open
Abstract
The ability to dynamically regulate, traffic, retain, and recycle proteins within the cell membrane is fundamental to life and central to the normal function of the heart. In the cardiomyocyte, these pathways are essential for the regulation of Ca(2+), both at the level of the plasma membrane, but also in local cellular domains. One intracellular pathway often overlooked in relation to cardiovascular Ca(2+) regulation and signaling is the endosome-based trafficking pathway. Highlighting its importance, this system and its molecular components are evolutionarily conserved across all metazoans. However, remarkably little is known of how endosome-based protein trafficking and recycling functions within mammalian cells systems, especially in the heart. As the endosomal system acts to regulate the expression and localization of membrane proteins central for cardiac Ca(2+) regulation, understanding the in vivo function of this system in the heart is critical. This review will focus on endosome-based protein trafficking in the heart in both health and disease with special emphasis for the role of endocytic regulatory proteins, C-terminal Eps15 homology domain-containing proteins (EHDs).
Collapse
Affiliation(s)
- Jerry Curran
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Michael A Makara
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Peter J Mohler
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Department of Internal Medicine, The Ohio State University Wexner Medical Center Columbus, OH, USA
| |
Collapse
|
35
|
Curran J, Makara MA, Little SC, Musa H, Liu B, Wu X, Polina I, Alecusan JS, Wright P, Li J, Billman GE, Boyden PA, Gyorke S, Band H, Hund TJ, Mohler PJ. EHD3-dependent endosome pathway regulates cardiac membrane excitability and physiology. Circ Res 2014; 115:68-78. [PMID: 24759929 DOI: 10.1161/circresaha.115.304149] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Cardiac function is dependent on the coordinate activities of membrane ion channels, transporters, pumps, and hormone receptors to tune the membrane electrochemical gradient dynamically in response to acute and chronic stress. Although our knowledge of membrane proteins has rapidly advanced during the past decade, our understanding of the subcellular pathways governing the trafficking and localization of integral membrane proteins is limited and essentially unstudied in vivo. In the heart, to our knowledge, there are no in vivo mechanistic studies that directly link endosome-based machinery with cardiac physiology. OBJECTIVE To define the in vivo roles of endosome-based cellular machinery for cardiac membrane protein trafficking, myocyte excitability, and cardiac physiology. METHODS AND RESULTS We identify the endosome-based Eps15 homology domain 3 (EHD3) pathway as essential for cardiac physiology. EHD3-deficient hearts display structural and functional defects including bradycardia and rate variability, conduction block, and blunted response to adrenergic stimulation. Mechanistically, EHD3 is critical for membrane protein trafficking, because EHD3-deficient myocytes display reduced expression/localization of Na/Ca exchanger and L-type Ca channel type 1.2 with a parallel reduction in Na/Ca exchanger-mediated membrane current and Cav1.2-mediated membrane current. Functionally, EHD3-deficient myocytes show increased sarcoplasmic reticulum [Ca], increased spark frequency, and reduced expression/localization of ankyrin-B, a binding partner for EHD3 and Na/Ca exchanger. Finally, we show that in vivo EHD3-deficient defects are attributable to cardiac-specific roles of EHD3 because mice with cardiac-selective EHD3 deficiency demonstrate both structural and electric phenotypes. CONCLUSIONS These data provide new insight into the critical role of endosome-based pathways in membrane protein targeting and cardiac physiology. EHD3 is a critical component of protein trafficking in heart and is essential for the proper membrane targeting of select cellular proteins that maintain excitability.
Collapse
Affiliation(s)
- Jerry Curran
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.).
| | - Michael A Makara
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Sean C Little
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Hassan Musa
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Bin Liu
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Xiangqiong Wu
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Iuliia Polina
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Joseph S Alecusan
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Patrick Wright
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Jingdong Li
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - George E Billman
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Penelope A Boyden
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Sandor Gyorke
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Hamid Band
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Thomas J Hund
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| | - Peter J Mohler
- From The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (J.C., M.A.M., S.C.L., H.M., B.L., X.W., I.P., J.S.A., P.W., J.L., G.E.B., S.G., T.J.H., P.J.M.); Departments of Internal Medicine (P.J.M.) and Physiology and Cell Biology (J.C., M.A.M., S.C.L., H.M., B.L., P.W., G.E.B., S.G., P.J.M.), The Ohio State University, Columbus; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus (T.J.H.); Department of Pharmacology and Center for Molecular Therapeutics, Columbia University Medical Center, New York, NY (P.A.B.); and The Eppley Institute and UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha (H.B.)
| |
Collapse
|
36
|
Posey AD, Swanson KE, Alvarez MG, Krishnan S, Earley JU, Band H, Pytel P, McNally EM, Demonbreun AR. EHD1 mediates vesicle trafficking required for normal muscle growth and transverse tubule development. Dev Biol 2014; 387:179-90. [PMID: 24440153 DOI: 10.1016/j.ydbio.2014.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 01/03/2023]
Abstract
EHD proteins have been implicated in intracellular trafficking, especially endocytic recycling, where they mediate receptor and lipid recycling back to the plasma membrane. Additionally, EHDs help regulate cytoskeletal reorganization and induce tubule formation. It was previously shown that EHD proteins bind directly to the C2 domains in myoferlin, a protein that regulates myoblast fusion. Loss of myoferlin impairs normal myoblast fusion leading to smaller muscles in vivo but the intracellular pathways perturbed by loss of myoferlin function are not well known. We now characterized muscle development in EHD1-null mice. EHD1-null myoblasts display defective receptor recycling and mislocalization of key muscle proteins, including caveolin-3 and Fer1L5, a related ferlin protein homologous to myoferlin. Additionally, EHD1-null myoblast fusion is reduced. We found that loss of EHD1 leads to smaller muscles and myofibers in vivo. In wildtype skeletal muscle EHD1 localizes to the transverse tubule (T-tubule), and loss of EHD1 results in overgrowth of T-tubules with excess vesicle accumulation in skeletal muscle. We provide evidence that tubule formation in myoblasts relies on a functional EHD1 ATPase domain. Moreover, we extended our studies to show EHD1 regulates BIN1 induced tubule formation. These data, taken together and with the known interaction between EHD and ferlin proteins, suggests that the EHD proteins coordinate growth and development likely through mediating vesicle recycling and the ability to reorganize the cytoskeleton.
Collapse
Affiliation(s)
- Avery D Posey
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Kaitlin E Swanson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Manuel G Alvarez
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Swathi Krishnan
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Judy U Earley
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Hamid Band
- Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Elizabeth M McNally
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
37
|
Simone LC, Naslavsky N, Caplan S. Scratching the surface: actin' and other roles for the C-terminal Eps15 homology domain protein, EHD2. Histol Histopathol 2013; 29:285-92. [PMID: 24347515 DOI: 10.14670/hh-29.285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The C-terminal Eps15 homology domain-containing (EHD) proteins participate in multiple aspects of endocytic membrane trafficking. Of the four mammalian EHD proteins, EHD2 appears to be the most disparate, both in terms of sequence homology, and in subcellular localization/function. Since its initial description as a plasma membrane-associated protein, the precise function of EHD2 has remained enigmatic. Various reports have suggested roles for EHD2 at the plasma membrane, within the endocytic transport system, and even in the nucleus. For example, EHD2 facilitates membrane fusion/repair in muscle cells. Recently the focus has shifted to the role of EHD2 in regulating caveolae. Indeed, EHD2 is highly expressed in tissues rich in caveolae, including fat, muscle and blood vessels. This review highlights cumulative evidence linking EHD2 to actin-rich structures at the plasma membrane, where the plasma membrane-associated phospholipid phosphatidylinositol 4,5-bisphosphate controls EHD2 recruitment. Herein we examine the key pathways where EHD2 might function, and address its potential involvement in these processes.
Collapse
Affiliation(s)
- Laura C Simone
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
38
|
Waxmonsky NC, Conner SD. Αvβ3-integrin-mediated adhesion is regulated through an AAK1L- and EHD3-dependent rapid-recycling pathway. J Cell Sci 2013; 126:3593-601. [PMID: 23781025 DOI: 10.1242/jcs.122465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein transport through the endosome is critical for maintaining proper integrin cell surface integrin distribution to support cell adhesion, motility and viability. Here we employ a live-cell imaging approach to evaluate the relationship between integrin function and transport through the early endosome. We discovered that two early endosome factors, AAK1L and EHD3, are critical for αvβ3-integrin-mediated cell adhesion in HeLa cells. siRNA-mediated depletion of either factor delays short-loop β3 integrin recycling from the early endosome back to the cell surface. Total internal reflection fluorescence-based colocalization analysis reveals that β3 integrin transits AAK1L- and EHD3-positive endosomes near the cell surface, a subcellular location consistent with a rapid-recycling role for both factors. Moreover, structure-function analysis reveals that AAK1L kinase activity, as well as its C-terminal domain, is essential for cell adhesion maintenance. Taken together, these data reveal an important role for AAK1L and EHD3 in maintaining cell viability and adhesion by promoting αvβ3 integrin rapid recycling from the early endosome.
Collapse
Affiliation(s)
- Nicole C Waxmonsky
- Department of Genetics, Cell Biology and Development, The University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
39
|
DeGrande ST, Little SC, Nixon DJ, Wright P, Snyder J, Dun W, Murphy N, Kilic A, Higgins R, Binkley PF, Boyden PA, Carnes CA, Anderson ME, Hund TJ, Mohler PJ. Molecular mechanisms underlying cardiac protein phosphatase 2A regulation in heart. J Biol Chem 2013; 288:1032-46. [PMID: 23204520 PMCID: PMC3542989 DOI: 10.1074/jbc.m112.426957] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/21/2012] [Indexed: 11/06/2022] Open
Abstract
Kinase/phosphatase balance governs cardiac excitability in health and disease. Although detailed mechanisms for cardiac kinase regulation are established, far less is known regarding cardiac protein phosphatase 2A (PP2A) regulation. This is largely due to the complexity of the PP2A holoenzyme structure (combinatorial assembly of three subunit enzyme from >17 subunit genes) and the inability to segregate "global" PP2A function from the activities of multiple "local" holoenzyme populations. Here we report that PP2A catalytic, regulatory, and scaffolding subunits are tightly regulated at transcriptional, translational, and post-translational levels to tune myocyte function at base line and in disease. We show that past global read-outs of cellular PP2A activity more appropriately represent the collective activity of numerous individual PP2A holoenzymes, each displaying a specific subcellular localization (dictated by select PP2A regulatory subunits) as well as local specific post-translational catalytic subunit methylation and phosphorylation events that regulate local and rapid holoenzyme assembly/disassembly (via leucine carboxymethyltransferase 1/phosphatase methylesterase 1 (LCMT-1/PME-1). We report that PP2A subunits are selectively regulated between human and animal models, across cardiac chambers, and even within specific cardiac cell types. Moreover, this regulation can be rapidly tuned in response to cellular activation. Finally, we report that global PP2A is altered in human and experimental models of heart disease, yet each pathology displays its own distinct molecular signature though specific PP2A subunit modulatory events. These new data provide an initial view into the signaling pathways that govern PP2A function in heart but also establish the first step in defining specific PP2A regulatory targets in health and disease.
Collapse
Affiliation(s)
- Sean T. DeGrande
- From The Dorothy M. Davis Heart and Lung Research Institute
- Department of Internal Medicine, Division of Cardiovascular Medicine
- the Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, and
| | - Sean C. Little
- From The Dorothy M. Davis Heart and Lung Research Institute
| | - Derek J. Nixon
- From The Dorothy M. Davis Heart and Lung Research Institute
| | - Patrick Wright
- From The Dorothy M. Davis Heart and Lung Research Institute
| | - Jedidiah Snyder
- From The Dorothy M. Davis Heart and Lung Research Institute
- Department of Internal Medicine, Division of Cardiovascular Medicine
- College of Engineering, Department of Biomedical Engineering, and
| | - Wen Dun
- the Department of Pharmacology, Columbia University, New York, New York 10032
| | | | - Ahmet Kilic
- From The Dorothy M. Davis Heart and Lung Research Institute
- Division of Cardiac Surgery, The Ohio State University Wexner Medical Center
| | - Robert Higgins
- From The Dorothy M. Davis Heart and Lung Research Institute
- Division of Cardiac Surgery, The Ohio State University Wexner Medical Center
| | - Philip F. Binkley
- From The Dorothy M. Davis Heart and Lung Research Institute
- Department of Internal Medicine, Division of Cardiovascular Medicine
| | - Penelope A. Boyden
- the Department of Pharmacology, Columbia University, New York, New York 10032
| | - Cynthia A. Carnes
- From The Dorothy M. Davis Heart and Lung Research Institute
- The College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - Mark E. Anderson
- the Department of Internal Medicine, Division of Cardiovascular Medicine and
- the Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, and
| | - Thomas J. Hund
- From The Dorothy M. Davis Heart and Lung Research Institute
- Department of Internal Medicine, Division of Cardiovascular Medicine
- College of Engineering, Department of Biomedical Engineering, and
| | - Peter J. Mohler
- From The Dorothy M. Davis Heart and Lung Research Institute
- Department of Internal Medicine, Division of Cardiovascular Medicine
- Department of Physiology and Cell Biology
| |
Collapse
|
40
|
Kline CF, Mohler PJ. Evolving form to fit function: cardiomyocyte intercalated disc and transverse-tubule membranes. CURRENT TOPICS IN MEMBRANES 2013; 72:121-58. [PMID: 24210429 DOI: 10.1016/b978-0-12-417027-8.00004-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The vertebrate cardiac myocyte has evolved a highly organized cellular membrane architecture and cell-cell contacts in order to effectively transmit precisely timed and homogeneous depolarizing waves without failure (>2 billion times/human life span). Two unique specialized membrane domains, the intercalated disc and the transverse tubule (T-tubule), function to ensure the rapid and coordinated propagation of the action potential throughout the heart. Based on their critical roles in structure, signaling, and electric inter- and intracellular communication, it is not surprising that dysfunction in these membrane structures is associated with aberrant vertebrate physiology, resulting in potentially fatal congenital and acquired disease. This chapter will review the fundamental components of cardiomyocyte intercalated disc and transverse-tubule membranes with a focus on linking dysfunction in these membranes with human cardiovascular disease.
Collapse
Affiliation(s)
- Crystal F Kline
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | |
Collapse
|
41
|
Bennett V, Lorenzo DN. Spectrin- and Ankyrin-Based Membrane Domains and the Evolution of Vertebrates. CURRENT TOPICS IN MEMBRANES 2013; 72:1-37. [DOI: 10.1016/b978-0-12-417027-8.00001-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Gatti S, Leo C, Gallo S, Sala V, Bucci E, Natale M, Cantarella D, Medico E, Crepaldi T. Gene expression profiling of HGF/Met activation in neonatal mouse heart. Transgenic Res 2012; 22:579-93. [DOI: 10.1007/s11248-012-9667-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 10/11/2012] [Indexed: 12/15/2022]
|
43
|
Williams R. Circulation Research
“In This Issue” Anthology. Circ Res 2012. [DOI: 10.1161/res.0b013e31826f7938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Circulation Research
Thematic Synopsis Cardiac Myocyte Biology and Function. Circ Res 2012. [DOI: 10.1161/circresaha.112.280974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Mate SE, Van Der Meulen JH, Arya P, Bhattacharyya S, Band H, Hoffman EP. Eps homology domain endosomal transport proteins differentially localize to the neuromuscular junction. Skelet Muscle 2012; 2:19. [PMID: 22974368 PMCID: PMC3541266 DOI: 10.1186/2044-5040-2-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 07/10/2012] [Indexed: 01/19/2023] Open
Abstract
Background Recycling of endosomes is important for trafficking and maintenance of proteins at the neuromuscular junction (NMJ). We have previously shown high expression of the endocytic recycling regulator Eps15 homology domain-containing (EHD)1 proteinin the Torpedo californica electric organ, a model tissue for investigating a cholinergic synapse. In this study, we investigated the localization of EHD1 and its paralogs EHD2, EHD3, and EHD4 in mouse skeletal muscle, and assessed the morphological changes in EHD1−/− NMJs. Methods Localization of the candidate NMJ protein EHD1 was assessed by confocal microscopy analysis of whole-mount mouse skeletal muscle fibers after direct gene transfer and immunolabeling. The potential function of EHD1 was assessed by specific force measurement and α-bungarotoxin-based endplate morphology mapping in EHD1−/− mouse skeletal muscle. Results Endogenous EHD1 localized to primary synaptic clefts of murine NMJ, and this localization was confirmed by expression of recombinant green fluorescent protein labeled-EHD1 in murine skeletal muscle in vivo. EHD1−/− mouse skeletal muscle had normal histology and NMJ morphology, and normal specific force generation during muscle contraction. The EHD 1–4 proteins showed differential localization in skeletal muscle: EHD2 to muscle vasculature, EHD3 to perisynaptic regions, and EHD4 to perinuclear regions and to primary synaptic clefts, but at lower levels than EHD1. Additionally, specific antibodies raised against mammalian EHD1-4 recognized proteins of the expected mass in the T. californica electric organ. Finally, we found that EHD4 expression was more abundant in EHD1−/− mouse skeletal muscle than in wild-type skeletal muscle. Conclusion EHD1 and EHD4 localize to the primary synaptic clefts of the NMJ. Lack of obvious defects in NMJ structure and muscle function in EHD1−/− muscle may be due to functional compensation by other EHD paralogs.
Collapse
Affiliation(s)
- Suzanne E Mate
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Kashef F, Li J, Wright P, Snyder J, Suliman F, Kilic A, Higgins RSD, Anderson ME, Binkley PF, Hund TJ, Mohler PJ. Ankyrin-B protein in heart failure: identification of a new component of metazoan cardioprotection. J Biol Chem 2012; 287:30268-81. [PMID: 22778271 DOI: 10.1074/jbc.m112.368415] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ankyrins (ankyrin-R, -B, and -G) are adapter proteins linked with defects in metazoan physiology. Ankyrin-B (encoded by ANK2) loss-of-function mutations are directly associated with human cardiovascular phenotypes including sinus node disease, atrial fibrillation, ventricular tachycardia, and sudden cardiac death. Despite the link between ankyrin-B dysfunction and monogenic disease, there are no data linking ankyrin-B regulation with common forms of human heart failure. Here, we report that ankyrin-B levels are altered in both ischemic and non-ischemic human heart failure. Mechanistically, we demonstrate that cardiac ankyrin-B levels are tightly regulated downstream of reactive oxygen species, intracellular calcium, and the calcium-dependent protease calpain, all hallmarks of human myocardial injury and heart failure. Surprisingly, β(II)-spectrin, previously thought to mediate ankyrin-dependent modulation in the nervous system and heart, is not coordinately regulated with ankyrin-B or its downstream partners. Finally, our data implicate ankyrin-B expression as required for vertebrate myocardial protection as hearts deficient in ankyrin-B show increased cardiac damage and impaired function relative to wild-type mouse hearts following ischemia reperfusion. In summary, our findings provide the data of ankyrin-B regulation in human heart failure, provide insight into candidate pathways for ankyrin-B regulation in acquired human cardiovascular disease, and surprisingly, implicate ankyrin-B as a molecular component for cardioprotection following ischemia.
Collapse
Affiliation(s)
- Farshid Kashef
- Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Smith S, Curran J, Hund TJ, Mohler PJ. Defects in cytoskeletal signaling pathways, arrhythmia, and sudden cardiac death. Front Physiol 2012; 3:122. [PMID: 22586405 PMCID: PMC3343379 DOI: 10.3389/fphys.2012.00122] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 04/12/2012] [Indexed: 11/25/2022] Open
Abstract
Ankyrin polypeptides are cellular adapter proteins that tether integral membrane proteins to the cytoskeleton in a host of human organs. Initially identified as integral components of the cytoskeleton in erythrocytes, a recent explosion in ankyrin research has demonstrated that these proteins play prominent roles in cytoskeletal signaling pathways and membrane protein trafficking/regulation in a variety of excitable and non-excitable cells including heart and brain. Importantly, ankyrin research has translated from bench to bedside with the discovery of human gene variants associated with ventricular arrhythmias that alter ankyrin–based pathways. Ankyrin polypeptides have also been found to play an instrumental role in various forms of sinus node disease and atrial fibrillation (AF). Mouse models of ankyrin-deficiency have played fundamental roles in the translation of ankyrin-based research to new clinical understanding of human sinus node disease, AF, and ventricular tachycardia.
Collapse
Affiliation(s)
- Sakima Smith
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center Columbus, OH, USA
| | | | | | | |
Collapse
|
48
|
Gudmundsson H, Curran J, Kashef F, Snyder JS, Smith SA, Vargas-Pinto P, Bonilla IM, Weiss RM, Anderson ME, Binkley P, Felder RB, Carnes CA, Band H, Hund TJ, Mohler PJ. Differential regulation of EHD3 in human and mammalian heart failure. J Mol Cell Cardiol 2012; 52:1183-90. [PMID: 22406195 DOI: 10.1016/j.yjmcc.2012.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/12/2012] [Accepted: 02/23/2012] [Indexed: 12/25/2022]
Abstract
Electrical and structural remodeling during the progression of cardiovascular disease is associated with adverse outcomes subjecting affected patients to overt heart failure (HF) and/or sudden death. Dysfunction in integral membrane protein trafficking has long been linked with maladaptive electrical remodeling. However, little is known regarding the molecular identity or function of these intracellular targeting pathways in the heart. Eps15 homology domain-containing (EHD) gene products (EHD1-4) are polypeptides linked with endosomal trafficking, membrane protein recycling, and lipid homeostasis in a wide variety of cell types. EHD3 was recently established as a critical mediator of membrane protein trafficking in the heart. Here, we investigate the potential link between EHD3 function and heart disease. Using four different HF models including ischemic rat heart, pressure overloaded mouse heart, chronic pacing-induced canine heart, and non-ischemic failing human myocardium we provide the first evidence that EHD3 levels are consistently increased in HF. Notably, the expression of the Na/Ca exchanger (NCX1), targeted by EHD3 in heart is similarly elevated in HF. Finally, we identify a molecular pathway for EHD3 regulation in heart failure downstream of reactive oxygen species and angiotensin II signaling. Together, our new data identify EHD3 as a previously unrecognized component of the cardiac remodeling pathway.
Collapse
Affiliation(s)
- Hjalti Gudmundsson
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Curran J, Mohler PJ. Coordinating electrical activity of the heart: ankyrin polypeptides in human cardiac disease. Expert Opin Ther Targets 2011; 15:789-801. [PMID: 21457127 DOI: 10.1517/14728222.2011.575363] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Over the past ten years, ankyrin polypeptides have emerged as players in cardiac excitation-contraction coupling. Once thought to solely play a structural role, loss-of-function variants of genes encoding ankyrin polypeptides have highlighted how this protein mediates subcellular localization of various electrical components of the excitation-contraction coupling machinery. Evidence has revealed how disruption of this localization is the primary cause of various cardiomyopathies, ranging from long-QT syndrome 4, to sinus node disease, to more common forms of arrhythmias. AREAS COVERED The roles of ankyrin polypeptides in excitation-contraction coupling in the heart and the development of ankyrin-specific cardiomyopathies. How ankyrin polypeptides may be involved in structural and electrical remodeling of the heart, post-myocardial infarct. How ankyrin interactions with membrane-bound ion channels may regulate these channels' response to stimuli. New data, which offers the potential for unique therapies, for not only combating heart disease, but also for wider applications to various disease states. EXPERT OPINION The ankyrin family of adapter proteins is emerging as an intimate player in cardiac excitation-contraction coupling. Until recently, these proteins have gone largely unappreciated for their importance in proper cardiac function. New insights into how these proteins function within the heart are offering potentially new avenues for therapies against cardiomyopathy.
Collapse
Affiliation(s)
- Jerry Curran
- The Ohio State University, The Dorothy M. Davis Heart and Lung Research Institute, Columbus, 43210, USA.
| | | |
Collapse
|
50
|
George M, Rainey MA, Naramura M, Foster KW, Holzapfel MS, Willoughby LL, Ying G, Goswami RM, Gurumurthy CB, Band V, Satchell SC, Band H. Renal thrombotic microangiopathy in mice with combined deletion of endocytic recycling regulators EHD3 and EHD4. PLoS One 2011; 6:e17838. [PMID: 21408024 PMCID: PMC3052385 DOI: 10.1371/journal.pone.0017838] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Accepted: 02/15/2011] [Indexed: 11/19/2022] Open
Abstract
Eps15 Homology Domain-containing 3 (EHD3), a member of the EHD protein family that regulates endocytic recycling, is the first protein reported to be specifically expressed in the glomerular endothelium in the kidney; therefore we generated Ehd3(-/-) mice and assessed renal development and pathology. Ehd3(-/-) animals showed no overt defects, and exhibited no proteinuria or glomerular pathology. However, as the expression of EHD4, a related family member, was elevated in the glomerular endothelium of Ehd3(-/-) mice and suggested functional compensation, we generated and analyzed Ehd3(-/-); Ehd4(-/-) mice. These mice were smaller, possessed smaller and paler kidneys, were proteinuric and died between 3-24 weeks of age. Detailed analyses of Ehd3(-/-); Ehd4(-/-) kidneys demonstrated thrombotic microangiopathy (TMA)-like glomerular lesions including thickening and duplication of glomerular basement membrane, endothelial swelling and loss of fenestrations. Other changes included segmental podocyte foot process effacement, mesangial interposition, and abnormal podocytic and mesangial marker expression. The glomerular lesions observed were strikingly similar to those seen in human pre-eclampsia and mouse models of reduced VEGF expression. As altered glomerular endothelial VEGFR2 expression and localization and increased apoptosis was observed in the absence of EHD3 and EHD4, we propose that EHD-mediated endocytic traffic of key surface receptors such as VEGFR2 is essential for physiological control of glomerular function. Furthermore, Ehd3(-/-); Ehd4(-/-) mice provide a unique model to elucidate mechanisms of glomerular endothelial injury which is observed in a wide variety of human renal and extra-renal diseases.
Collapse
Affiliation(s)
- Manju George
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MG); (MN); (HB)
| | - Mark A. Rainey
- Department of Pharmacology, Creighton University, Omaha, Nebraska, United States of America
| | - Mayumi Naramura
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MG); (MN); (HB)
| | - Kirk W. Foster
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Melissa S. Holzapfel
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Laura L. Willoughby
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - GuoGuang Ying
- Oncology Central Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Rasna M. Goswami
- Abbott Laboratories, Abbott Park, Illinois, United States of America
| | - Channabasavaiah B. Gurumurthy
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | | | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Departments of Biochemistry and Molecular Biology, and Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (MG); (MN); (HB)
| |
Collapse
|