1
|
Niu N, Miao H, Ren H. Transcriptome Analysis of Myocardial Ischemic-Hypoxic Injury in Rats and Hypoxic H9C2 Cells. ESC Heart Fail 2024; 11:3775-3795. [PMID: 39010664 PMCID: PMC11631282 DOI: 10.1002/ehf2.14903] [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: 06/26/2023] [Revised: 04/18/2024] [Accepted: 05/24/2024] [Indexed: 07/17/2024] Open
Abstract
AIMS This study aimed to address inconsistencies in results between the H9C2 myocardial hypoxia (MH) cell line and myocardial infarction (MI) rat models used in MI research. We identified differentially expressed genes (DEGs) and underlying molecular mechanisms using RNA sequencing technology. METHODS RNA sequencing was used to analyse DEGs in MI rat tissues and H9C2 cells exposed to hypoxia for 24 h. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were used to identify key biological processes and pathways. Weighted correlation network analysis [weighted gene co-expression network analysis (WGCNA)] was used to construct gene co-expression networks, and hub genes were compared with published MI datasets [Gene Expression Omnibus (GEO)] for target identification. RESULTS GO analysis revealed enrichment of immune inflammation and mitochondrial respiration processes among 5139 DEGs in MI tissues and 2531 in H9C2 cells. KEGG analysis identified 537 overlapping genes associated with metabolism and oxidative stress pathways. Cross-analyses using the published GSE35088 and GSE47495 datasets identified 40 and 16 overlapping genes, respectively, with nine genes overlapping across all datasets and our models. WGCNA identified a key module in the MI model enriched for mRNA processing and protein binding. GO analysis revealed enrichment of mRNA processing, protein binding and mitochondrial respiratory chain complex I assembly in MI and H9C2 MH models. Five relevant hub genes were identified via a cross-analysis between the 92 hub genes that showed a common expression trend in both models. CONCLUSIONS This study reveals both shared and distinct transcriptomic responses in the MI and H9C2 models, highlighting the importance of model selection for studying myocardial ischaemia and hypoxia.
Collapse
Affiliation(s)
- Nan Niu
- Department of Cardiovascular MedicinePeople's Hospital of Ningxia Hui Autonomous RegionYinchuanChina
| | - Huangtai Miao
- Coronary Heart Disease Center,Beijing Anzhen Hospital, Capital Medical UniversityBeijingChina
| | - Hongmei Ren
- Department of Cardiovascular MedicinePeople's Hospital of Ningxia Hui Autonomous RegionYinchuanChina
| |
Collapse
|
2
|
Schaiter A, Hentschel A, Kleefeld F, Schuld J, Umathum V, Procida-Kowalski T, Nelke C, Roth A, Hahn A, Krämer HH, Ruck T, Horvath R, van der Ven PFM, Bartkuhn M, Roos A, Schänzer A. Molecular composition of skeletal muscle in infants and adults: a comparative proteomic and transcriptomic study. Sci Rep 2024; 14:22965. [PMID: 39362957 PMCID: PMC11450201 DOI: 10.1038/s41598-024-74913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024] Open
Abstract
To gain a deeper understanding of skeletal muscle function in younger age and aging in elderly, identification of molecular signatures regulating these functions under physiological conditions is needed. Although molecular studies of healthy muscle have been conducted on adults and older subjects, there is a lack of research on infant muscle in terms of combined morphological, transcriptomic and proteomic profiles. To address this gap of knowledge, we performed RNA sequencing (RNA-seq), tandem mass spectrometry (LC-MS/MS), morphometric analysis and assays for mitochondrial maintenance in skeletal muscle biopsies from both, infants aged 4-28 months and adults aged 19-65 years. We identified differently expressed genes (DEGs) and differentially expressed proteins (DEPs) in adults compared to infants. The down-regulated genes in adults were associated with functional terms primarily related to sarcomeres, cellular maintenance, and metabolic, immunological and developmental processes. Thus, our study indicates age-related differences in the molecular signatures and associated functions of healthy skeletal muscle. Moreover, the findings assert that processes previously associated solely with aging are indeed part of development and healthy aging. Hence, combined findings of this study also indicate that age-dependent controls are crucial in muscle disease studies, as otherwise the comparative results may not be reliable.
Collapse
Affiliation(s)
| | - Andreas Hentschel
- Leibnitz Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
| | - Felix Kleefeld
- Department of Clinical Neurosciences, School of Clinical Medicine, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Schuld
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Vincent Umathum
- Institute of Neuropathology, Justus-Liebig University, Giessen, Germany
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | | | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Angela Roth
- Institute of Neuropathology, Justus-Liebig University, Giessen, Germany
| | - Andreas Hahn
- Department of Pediatric Neurology, Justus-Liebig University Giessen, Giessen, Germany
| | - Heidrun H Krämer
- Department of Neurology, Justus-Liebig University Giessen, Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, School of Clinical Medicine, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Marek Bartkuhn
- Institute for Lung Health (ILH), Justus-Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Roos
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University, Giessen, Germany.
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
3
|
Yan Y, Qin X, Zheng Y, Jin T, Hu Y, An Q, Leng B. Decreased PDLIM1 expression in endothelial cells contributes to the development of intracranial aneurysm. Vasc Med 2024; 29:5-16. [PMID: 38334094 DOI: 10.1177/1358863x231218210] [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] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Intracranial aneurysm (IA) is a common vascular enlargement that occurs in the wall of cerebral vessels and frequently leads to fatal subarachnoid hemorrhage. PDZ and LIM domain protein 1 (PDLIM1) is a cytoskeletal protein that functions as a platform for multiple protein complex formation. However, whether PDLIM is involved in the pathogenesis of IA remains poorly understood. METHODS Loss-of-function and gain-of-function strategies were employed to determine the in vitro roles of PDLIM1 in vascular endothelial cells (VECs). A rat model of IA was generated to study the role of PDLIM1 in vivo. Gene expression profiling, Western blotting, and dual luciferase reporter assays were performed to uncover the underlying cellular mechanism. Clinical IA samples were used to determine the expression of PDLIM1 and its downstream signaling molecules. RESULTS PDLIM1 expression was reduced in the endothelial cells of IA and was regulated by Yes-associated protein 1 (YAP1). Genetic silencing of PDLIM1 inhibited the viability, migratory ability, and tube formation ability of VECs. Opposite results were obtained by ectopic expression of PDLIM1. Additionally, PDLIM1 overexpression mitigated IA in vivo. Mechanistic investigations revealed that PDLIM1 promoted the transcriptional activity of β-catenin and induced the expression of v-myc myelocytomatosis viral oncogene homolog (MYC) and cyclin D1 (CCND1). In clinical settings, reduced expression of PDLIM1 and β-catenin downstream target genes was observed in human IA samples. CONCLUSION Our study indicates that YAP1-dependent expression of PDLIM1 can inhibit IA development by modulating the activity of the Wnt/β-catenin signaling pathway and that PDLIM1 deficiency in VECs may represent a potential marker of aggressive disease.
Collapse
Affiliation(s)
- Yan Yan
- Department of Neurosurgery, The 1st Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanfeng Qin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongtao Zheng
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Jin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Leng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Lei Y, He L, Li Y, Hou J, Zhang H, Li G. PDLIM1 interacts with HK2 to promote gastric cancer progression through enhancing the Warburg effect via Wnt/β-catenin signaling. Cell Tissue Res 2024; 395:105-116. [PMID: 37930472 DOI: 10.1007/s00441-023-03840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
PDZ and LIM domain protein 1 (PDLIM1) is a cytoskeletal protein and is associated with the malignant pathological features of several tumors. However, the prognostic value of PDLIM1 and the molecular mechanisms by which it is involved in the metabolism and progression in gastric cancer (GC) are still unclear. The GEPIA database was used to predict the expression and prognosis of PDLIM1 in GC. qRT-PCR and western blot assays were applied to detect the mRNA and protein expression in GC tissues and cells. Loss- and gain-of-function experiments were performed to evaluate the biological role of PDLIM1 in GC cells. The Warburg effect was detected by a battery of glycolytic indicators. The interaction of PDLIM1 and hexokinase 2 (HK2) was determined by a co-immunoprecipitation assay. Furthermore, the modulatory effects of PDLIM1 and HK2 on Wnt/β-catenin signaling were assessed. The results showed that PDLIM1 expression was upregulated in GC tissues and cells and was associated with a poor prognosis for GC patients. PDLIM1 inhibition reduced GC cell proliferation, migration and invasion and promoted cell apoptosis. In the glucose deprivation (GLU-D) condition, the PDLIM1 level was reduced and PDLIM1 overexpression led to an increase in glycolysis. Besides, mechanistic investigation showed that PDLIM1 interacted with HK2 to mediate biological behaviors and the glycolysis of GC through Wnt/β-catenin signaling under glucose deprivation. In conclusion, PDLIM1 interacts with HK2 to promote gastric cancer progression by enhancing the Warburg effect via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yunpeng Lei
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Lirui He
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Yue Li
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Jianing Hou
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Haoran Zhang
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Guan Li
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
5
|
Fisher LAB, Schöck F. The unexpected versatility of ALP/Enigma family proteins. Front Cell Dev Biol 2022; 10:963608. [PMID: 36531944 PMCID: PMC9751615 DOI: 10.3389/fcell.2022.963608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
One of the most intriguing features of multicellular animals is their ability to move. On a cellular level, this is accomplished by the rearrangement and reorganization of the cytoskeleton, a dynamic network of filamentous proteins which provides stability and structure in a stationary context, but also facilitates directed movement by contracting. The ALP/Enigma family proteins are a diverse group of docking proteins found in numerous cellular milieus and facilitate these processes among others. In vertebrates, they are characterized by having a PDZ domain in combination with one or three LIM domains. The family is comprised of CLP-36 (PDLIM1), Mystique (PDLIM2), ALP (PDLIM3), RIL (PDLIM4), ENH (PDLIM5), ZASP (PDLIM6), and Enigma (PDLIM7). In this review, we will outline the evolution and function of their protein domains which confers their versatility. Additionally, we highlight their role in different cellular environments, focusing specifically on recent advances in muscle research using Drosophila as a model organism. Finally, we show the relevance of this protein family to human myopathies and the development of muscle-related diseases.
Collapse
|
6
|
Zhou JK, Fan X, Cheng J, Liu W, Peng Y. PDLIM1: Structure, function and implication in cancer. Cell Stress 2021; 5:119-127. [PMID: 34396044 PMCID: PMC8335553 DOI: 10.15698/cst2021.08.254] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/05/2023] Open
Abstract
PDLIM1, a member of the PDZ-LIM family, is a cytoskeletal protein and functions as a platform to form distinct protein complexes, thus participating in multiple physiological processes such as cytoskeleton regulation and synapse formation. Emerging evidence demonstrates that PDLIM1 is dysregualted in a variety of tumors and plays essential roles in tumor initiation and progression. In this review, we summarize the structure and function of PDLIM1, as well as its important roles in human cancers.
Collapse
Affiliation(s)
- Jian-Kang Zhou
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Fan
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Cheng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenrong Liu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Liu Y, Wang C, Cheng P, Zhang S, Zhou W, Xu Y, Xu H, Ji G. FHL1 Inhibits the Progression of Colorectal Cancer by Regulating the Wnt/β-Catenin Signaling Pathway. J Cancer 2021; 12:5345-5354. [PMID: 34335951 PMCID: PMC8317513 DOI: 10.7150/jca.60543] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/15/2021] [Indexed: 11/17/2022] Open
Abstract
Purpose: This study aims to explore the FHL1 expression level in colorectal cancer (CRC) patients, analyze its association with patient survival and investigate the role of FHL1 in CRC. Methods: We used secondary sequencing to profile mRNA expression in CRC tissue and corresponding adjacent normal tissue from four CRC patients. We focus on FHL1 and analyzed the association between its expression level and clinical indicators. Furthermore, we explored the functional role of FHL1 in colorectal cancer tumorigenesis by transfecting cells with siRNA or overexpression plasmids. Results: Hierarchical clustering revealed significantly differentially expressed mRNAs. FHL1 expression was significantly lower in CRC tissue than in adjacent normal tissue as well as in CRC cell lines relative to NCM460. Low FHL1 expression in CRC tissue correlated with poor patient survival. Our data demonstrated that overexpression of FHL1 inhibited the proliferation, colony formation potential, and expression of CdK4 and Cyclin D1, whereas ablating FHL1 promoted their proliferation and colony formation potential, suggesting that FHL1 acts as a tumor suppressor in CRC. Moreover, we showed that FHL1 inhibited the proliferation of colorectal cancer cells by negatively regulating the Wnt/β-catenin signaling pathway. Conclusion: FHL1 is a potential tumor suppressor gene in colorectal cancer, and regulation of the FHL1-Wnt/β-catenin pathway may be part of its antitumor mechanism.
Collapse
Affiliation(s)
- Yujing Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chunyan Wang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Peiqiu Cheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Shengan Zhang
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yangxian Xu
- Department of General Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
8
|
Deshmukh AS, Steenberg DE, Hostrup M, Birk JB, Larsen JK, Santos A, Kjøbsted R, Hingst JR, Schéele CC, Murgia M, Kiens B, Richter EA, Mann M, Wojtaszewski JFP. Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training. Nat Commun 2021; 12:304. [PMID: 33436631 PMCID: PMC7803955 DOI: 10.1038/s41467-020-20556-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
Skeletal muscle conveys several of the health-promoting effects of exercise; yet the underlying mechanisms are not fully elucidated. Studying skeletal muscle is challenging due to its different fiber types and the presence of non-muscle cells. This can be circumvented by isolation of single muscle fibers. Here, we develop a workflow enabling proteomics analysis of pools of isolated muscle fibers from freeze-dried human muscle biopsies. We identify more than 4000 proteins in slow- and fast-twitch muscle fibers. Exercise training alters expression of 237 and 172 proteins in slow- and fast-twitch muscle fibers, respectively. Interestingly, expression levels of secreted proteins and proteins involved in transcription, mitochondrial metabolism, Ca2+ signaling, and fat and glucose metabolism adapts to training in a fiber type-specific manner. Our data provide a resource to elucidate molecular mechanisms underlying muscle function and health, and our workflow allows fiber type-specific proteomic analyses of snap-frozen non-embedded human muscle biopsies.
Collapse
Affiliation(s)
- A S Deshmukh
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - D E Steenberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - M Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J B Birk
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J K Larsen
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - A Santos
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - C C Schéele
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - M Murgia
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - B Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - E A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - M Mann
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - J F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
9
|
Dhanda AS, Yang D, Kooner A, Guttman JA. Distribution of PDLIM1 at actin-rich structures generated by invasive and adherent bacterial pathogens. Anat Rec (Hoboken) 2020; 304:919-938. [PMID: 33022122 DOI: 10.1002/ar.24523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022]
Abstract
The enteric bacterial pathogens Listeria monocytogenes (Listeria) and enteropathogenic Escherichia coli (EPEC) remodel the eukaryotic actin cytoskeleton during their disease processes. Listeria generate slender actin-rich comet/rocket tails to move intracellularly, and later, finger-like membrane protrusions to spread amongst host cells. EPEC remain extracellular, but generate similar actin-rich membranous protrusions (termed pedestals) to move atop the host epithelia. These structures are crucial for disease as diarrheal (and systemic) infections are significantly abrogated during infections with mutant strains that are unable to generate the structures. The current repertoire of host components enriched within these structures is vast and diverse. In this protein catalog, we and others have found that host actin crosslinkers, such as palladin and α-actinin-1, are routinely exploited. To expand on this list, we set out to investigate the distribution of PDLIM1, a scaffolding protein and binding partner of palladin and α-actinin-1, during bacterial infections. We show that PDLIM1 localizes to the site of initial Listeria entry into cells. Following this, PDLIM1 localizes to actin filament clouds surrounding immotile bacteria, and then colocalizes with actin once the comet/rocket tails are generated. Unlike palladin or α-actinin-1, PDLIM1 is maintained within the actin-rich core of membrane protrusions. Conversely, α-actinin-1, but not PDLIM1 (or palladin), is enriched at the membrane invagination that internalizes the Listeria-containing membrane protrusion. We also show that PDLIM1 is a component of the EPEC pedestal core and that its recruitment is dependent on the bacterial effector Tir. Our findings highlight PDLIM1 as another protein present within pathogen-induced actin-rich structures.
Collapse
Affiliation(s)
- Aaron S Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Diana Yang
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Avneen Kooner
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
10
|
Huang Z, Zhou JK, Wang K, Chen H, Qin S, Liu J, Luo M, Chen Y, Jiang J, Zhou L, Zhu L, He J, Li J, Pu W, Gong Y, Li J, Ye Q, Dong D, Hu H, Zhou Z, Dai L, Huang C, Wei X, Peng Y. PDLIM1 Inhibits Tumor Metastasis Through Activating Hippo Signaling in Hepatocellular Carcinoma. Hepatology 2020; 71:1643-1659. [PMID: 31509262 DOI: 10.1002/hep.30930] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 09/02/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Tumor metastasis is a major factor of high recurrence and mortality in hepatocellular carcinoma (HCC), but its underlying mechanism remains elusive. We report that PDZ and LIM domain protein 1 (PDLIM1) is significantly down-regulated in metastatic human HCC tissues, which predicts unfavorable prognosis, suggesting that PDLIM1 may play an important inhibitory role during HCC metastasis. APPROACH AND RESULTS Functional studies indicate that PDLIM1 knockdown induces epithelial-to-mesenchymal transition (EMT) of HCC cells, elevates their invasive capacity, and promotes metastasis in vitro and in vivo, whereas overexpression of PDLIM1 exhibits opposite phenotypes. Mechanistically, PDLIM1 competitively binds to the cytoskeleton cross-linking protein alpha-actinin 4 (ACTN4), leading to the disassociation of ACTN4 from F-actin, thus preventing F-actin overgrowth. In contrast, loss of PDLIM1 induces excessive F-actin formation, resulting in dephosphorylation of large tumor suppressor kinase 1 and activation of Yes-associated protein, thereby promoting HCC metastasis. Moreover, Asn145 (N145) of PDLIM1 is critical for its interaction with ACTN4, and N145A mutation abolishes its regulatory function in Hippo signaling and HCC metastasis. CONCLUSIONS Our findings indicate that PDLIM1 suppresses HCC metastasis by modulating Hippo signaling, suggesting that PDLIM1 may be a potential prognostic marker for metastatic HCC.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jian-Kang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Kui Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Haining Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jiayang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lei Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Juan He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jiao Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenchen Pu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yanqiu Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jianbo Li
- Department of Liver Surgery and Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Ye
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dandan Dong
- Department of Pathology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
11
|
Li S, Liu C, Gu L, Wang L, Shang Y, Liu Q, Wan J, Shi J, Wang F, Xu Z, Ji G, Li W. Autophagy protects cardiomyocytes from the myocardial ischaemia-reperfusion injury through the clearance of CLP36. Open Biol 2017; 6:rsob.160177. [PMID: 27512143 PMCID: PMC5008017 DOI: 10.1098/rsob.160177] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of the death worldwide. An increasing number of studies have found that autophagy is involved in the progression or prevention of CVD. However, the precise mechanism of autophagy in CVD, especially the myocardial ischaemia-reperfusion injury (MI/R injury), is unclear and controversial. Here, we show that the cardiomyocyte-specific disruption of autophagy by conditional knockout of Atg7 leads to severe contractile dysfunction, myofibrillar disarray and vacuolar cardiomyocytes. A negative cytoskeleton organization regulator, CLP36, was found to be accumulated in Atg7-deficient cardiomyocytes. The cardiomyocyte-specific knockout of Atg7 aggravates the MI/R injury with cardiac hypertrophy, contractile dysfunction, myofibrillar disarray and severe cardiac fibrosis, most probably due to CLP36 accumulation in cardiomyocytes. Altogether, this work reveals autophagy may protect cardiomyocytes from the MI/R injury through the clearance of CLP36, and these findings define a novel relationship between autophagy and the regulation of stress fibre in heart.
Collapse
Affiliation(s)
- Shiguo Li
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lei Gu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lina Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yongliang Shang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qiong Liu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Junyi Wan
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jian Shi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zhiliang Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
12
|
Sharma P, Abbasi C, Lazic S, Teng ACT, Wang D, Dubois N, Ignatchenko V, Wong V, Liu J, Araki T, Tiburcy M, Ackerley C, Zimmermann WH, Hamilton R, Sun Y, Liu PP, Keller G, Stagljar I, Scott IC, Kislinger T, Gramolini AO. Evolutionarily conserved intercalated disc protein Tmem65 regulates cardiac conduction and connexin 43 function. Nat Commun 2015; 6:8391. [PMID: 26403541 DOI: 10.1038/ncomms9391] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
Membrane proteins are crucial to heart function and development. Here we combine cationic silica-bead coating with shotgun proteomics to enrich for and identify plasma membrane-associated proteins from primary mouse neonatal and human fetal ventricular cardiomyocytes. We identify Tmem65 as a cardiac-enriched, intercalated disc protein that increases during development in both mouse and human hearts. Functional analysis of Tmem65 both in vitro using lentiviral shRNA-mediated knockdown in mouse cardiomyocytes and in vivo using morpholino-based knockdown in zebrafish show marked alterations in gap junction function and cardiac morphology. Molecular analyses suggest that Tmem65 interaction with connexin 43 (Cx43) is required for correct localization of Cx43 to the intercalated disc, since Tmem65 deletion results in marked internalization of Cx43, a shorter half-life through increased degradation, and loss of Cx43 function. Our data demonstrate that the membrane protein Tmem65 is an intercalated disc protein that interacts with and functionally regulates ventricular Cx43.
Collapse
Affiliation(s)
- Parveen Sharma
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, Toronto, Ontario, Canada M5G 1L7
| | - Cynthia Abbasi
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, Toronto, Ontario, Canada M5G 1L7
| | - Savo Lazic
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Allen C T Teng
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, Toronto, Ontario, Canada M5G 1L7
| | - Dingyan Wang
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, Toronto, Ontario, Canada M5G 1L7
| | - Nicole Dubois
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Victoria Wong
- Departments of Molecular Genetics and Biochemistry, Donnelly Centre,, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Jun Liu
- Department of Mechanical and Industrial Engineering, Advanced Micro and Nanosystems Laboratory, University of Toronto, Toronto, Ontario, Canada M5S 3G8
| | - Toshiyuki Araki
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Malte Tiburcy
- Institute of Pharmacology, University Medical Center Göttingen and DZHK (German Center for Cardiovascular Research) partner site Göttingen, Göttingen 37075, Germany
| | - Cameron Ackerley
- The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Wolfram H Zimmermann
- Institute of Pharmacology, University Medical Center Göttingen and DZHK (German Center for Cardiovascular Research) partner site Göttingen, Göttingen 37075, Germany
| | - Robert Hamilton
- The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8.,Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada M5G 1L7
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, Advanced Micro and Nanosystems Laboratory, University of Toronto, Toronto, Ontario, Canada M5S 3G8
| | - Peter P Liu
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Gordon Keller
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Igor Stagljar
- Departments of Molecular Genetics and Biochemistry, Donnelly Centre,, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Ian C Scott
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8.,Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada M5G 1L7
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 2M9
| | - Anthony O Gramolini
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, Toronto, Ontario, Canada M5G 1L7.,Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada M5G 1L7
| |
Collapse
|
13
|
Transcriptional Response to Acute Thermal Exposure in Juvenile Chinook Salmon Determined by RNAseq. G3-GENES GENOMES GENETICS 2015; 5:1335-49. [PMID: 25911227 PMCID: PMC4502368 DOI: 10.1534/g3.115.017699] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thermal exposure is a serious and growing challenge facing fish species worldwide. Chinook salmon (Oncorhynchus tshawytscha) living in the southern portion of their native range are particularly likely to encounter warmer water due to a confluence of factors. River alterations have increased the likelihood that juveniles will be exposed to warm water temperatures during their freshwater life stage, which can negatively impact survival, growth, and development and pose a threat to dwindling salmon populations. To better understand how acute thermal exposure affects the biology of salmon, we performed a transcriptional analysis of gill tissue from Chinook salmon juveniles reared at 12° and exposed acutely to water temperatures ranging from ideal to potentially lethal (12° to 25°). Reverse-transcribed RNA libraries were sequenced on the Illumina HiSeq2000 platform and a de novo reference transcriptome was created. Differentially expressed transcripts were annotated using Blast2GO and relevant gene clusters were identified. In addition to a high degree of downregulation of a wide range of genes, we found upregulation of genes involved in protein folding/rescue, protein degradation, cell death, oxidative stress, metabolism, inflammation/immunity, transcription/translation, ion transport, cell cycle/growth, cell signaling, cellular trafficking, and structure/cytoskeleton. These results demonstrate the complex multi-modal cellular response to thermal stress in juvenile salmon.
Collapse
|
14
|
Zhang L, Chen X, Sharma P, Moon M, Sheftel AD, Dawood F, Nghiem MP, Wu J, Li RK, Gramolini AO, Sorensen PH, Penninger JM, Brumell JH, Liu PP. HACE1-dependent protein degradation provides cardiac protection in response to haemodynamic stress. Nat Commun 2014; 5:3430. [PMID: 24614889 PMCID: PMC3959209 DOI: 10.1038/ncomms4430] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 02/11/2014] [Indexed: 01/10/2023] Open
Abstract
The HECT E3 ubiquitin ligase HACE1
is a tumour suppressor known to regulate Rac1 activity under stress conditions. HACE1 is increased in the serum of patients
with heart failure. Here we show that HACE1 protects the heart under pressure stress by controlling
protein degradation. Hace1
deficiency in mice results in accelerated heart failure and increased mortality
under haemodynamic stress. Hearts from Hace1−/− mice
display abnormal cardiac hypertrophy, left ventricular dysfunction, accumulation of
LC3, p62 and ubiquitinated proteins enriched for
cytoskeletal species, indicating impaired autophagy. Our data suggest that
HACE1 mediates p62-dependent selective autophagic turnover
of ubiquitinated proteins by its ankyrin repeat domain through
protein–protein interaction, which is independent of its E3 ligase
activity. This would classify HACE1 as a dual-function E3 ligase. Our finding that
HACE1 has a protective
function in the heart in response to haemodynamic stress suggests that HACE1 may be a potential diagnostic and
therapeutic target for heart disease. HACE1 is an E3 ubiquitin ligase known to regulate various cell
biological processes. Here, Zhang et al. identify HACE1 as a protective factor in
the heart, demonstrating that HACE1 inhibits the development of heart failure in
response to haemodynamic stress by regulating protein degradation pathways.
Collapse
Affiliation(s)
- Liyong Zhang
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Xin Chen
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Parveen Sharma
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Mark Moon
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Alex D Sheftel
- University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7
| | - Fayez Dawood
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Mai P Nghiem
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Jun Wu
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Ren-Ke Li
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Anthony O Gramolini
- 1] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4 [2] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer Research Center, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1L3
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr Bohrgasse 3, A-1030 Vienna, Austria
| | - John H Brumell
- 1] Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [2] Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [3] Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1 × 8
| | - Peter P Liu
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4 [3] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [4] Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
15
|
Bertrand AT, Bönnemann CG, Bonne G. 199th ENMC international workshop: FHL1 related myopathies, June 7-9, 2013, Naarden, The Netherlands. Neuromuscul Disord 2014; 24:453-62. [PMID: 24613424 DOI: 10.1016/j.nmd.2014.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/04/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Anne T Bertrand
- Inserm, U974, Paris F-75013, France; Sorbonne Universités, UPMC Univ Paris 06, Myology Center of Research, UM76; CNRS FRE 3617, Institut de Myologie, Paris F-75013, France
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Gisèle Bonne
- Inserm, U974, Paris F-75013, France; Sorbonne Universités, UPMC Univ Paris 06, Myology Center of Research, UM76; CNRS FRE 3617, Institut de Myologie, Paris F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, U.F. Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, Paris F-75013, France.
| | | |
Collapse
|
16
|
Swelling-activated Cl− currents and intracellular CLC-3 are involved in proliferation of human pulmonary artery smooth muscle cells. J Hypertens 2014; 32:318-30. [DOI: 10.1097/hjh.0000000000000013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
17
|
Wang LL, Gu H, Fan Y, Zhang Y, Wu D, Miao JN, Huang TC, Li H, Yuan ZW. Up-regulated FHL1 expression maybe involved in the prognosis of Hirschsprung's disease. Int J Med Sci 2014; 11:262-7. [PMID: 24516350 PMCID: PMC3917115 DOI: 10.7150/ijms.7287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In a subset of patients with Hirschsprung's disease (HSCR), gastrointestinal motor dysfunction persisted long after surgical correction. Gastrointestinal motility is achieved through the coordinated activity of the enteric nervous system, interstitial cells of Cajal, and smooth muscle (SMC) cells. Inhibition of four-and-a-half LIM protein-1 (Fhl1) expression by siRNA significantly decreases pulmonary artery SMCs migration and proliferation. Furthermore when up-expressing FHL1 in atrial myocytes, K (+) current density markedly increases, therefore changing myocytes' response to an electrical stimulus. However whether FHL1 in colon SMCs (the final effector organ) influences intestinal motility in HSCR patients has not been clarified. METHODS FHL1 mRNA and protein expressions were analyzed in 32 HSCR colons and 4 normal colons. RESULTS Smooth muscle layers were thicken and disorganized in HSCR. FHL1 was expressed in the ganglion cells of the myenteric plexus, submucosa, as well as in the longitudinal and circular muscle layer of the ganglionic colon. FHL1 mRNA relative expression level in aganglionic colons was 1.06 ± 0.49 (ganglionic colon relative expression level was 1) (P=0.44). FHL1 protein gray level relative to GAPDH in normal colons was 0.83 ± 0.09. FHL1 expression level in ganglionic colon (1.66 ± 0.30) or aganglionic colon (1.81 ± 0.35) was significantly higher than that in normal colons (P=0.045 and P=0.041, respectively). Meanwhile, we found FHL1 expression in aganglionic colon was slightly stronger than that in ganglionic colon (P=0.036). CONCLUSION These data suggested that up-regulated FHL1 in smooth muscle in HSCR might be associated with intestinal wall remodeling in HSCR and might be one of the risk factors for gastrointestinal motor dysfunction.
Collapse
Affiliation(s)
- Li-Li Wang
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Hui Gu
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Yang Fan
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Yi Zhang
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Di Wu
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Jia-Ning Miao
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Tian-Chu Huang
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Hui Li
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| | - Zheng-Wei Yuan
- Key laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shen yang, 110004, P.R. China
| |
Collapse
|
18
|
Design and formulation of functional pluripotent stem cell-derived cardiac microtissues. Proc Natl Acad Sci U S A 2013; 110:E4698-707. [PMID: 24255110 DOI: 10.1073/pnas.1311120110] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Access to robust and information-rich human cardiac tissue models would accelerate drug-based strategies for treating heart disease. Despite significant effort, the generation of high-fidelity adult-like human cardiac tissue analogs remains challenging. We used computational modeling of tissue contraction and assembly mechanics in conjunction with microfabricated constraints to guide the design of aligned and functional 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues that we term cardiac microwires (CMWs). Miniaturization of the platform circumvented the need for tissue vascularization and enabled higher-throughput image-based analysis of CMW drug responsiveness. CMW tissue properties could be tuned using electromechanical stimuli and cell composition. Specifically, controlling self-assembly of 3D tissues in aligned collagen, and pacing with point stimulation electrodes, were found to promote cardiac maturation-associated gene expression and in vivo-like electrical signal propagation. Furthermore, screening a range of hPSC-derived cardiac cell ratios identified that 75% NKX2 Homeobox 5 (NKX2-5)+ cardiomyocytes and 25% Cluster of Differentiation 90 OR (CD90)+ nonmyocytes optimized tissue remodeling dynamics and yielded enhanced structural and functional properties. Finally, we demonstrate the utility of the optimized platform in a tachycardic model of arrhythmogenesis, an aspect of cardiac electrophysiology not previously recapitulated in 3D in vitro hPSC-derived cardiac microtissue models. The design criteria identified with our CMW platform should accelerate the development of predictive in vitro assays of human heart tissue function.
Collapse
|
19
|
Wang L, Miao J, Li L, Wu D, Zhang Y, Peng Z, Zhang L, Yuan Z, Sun K. Identification of an FHL1 protein complex containing gamma-actin and non-muscle myosin IIB by analysis of protein-protein interactions. PLoS One 2013; 8:e79551. [PMID: 24265776 PMCID: PMC3827166 DOI: 10.1371/journal.pone.0079551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 09/24/2013] [Indexed: 12/24/2022] Open
Abstract
FHL1 is multifunctional and serves as a modular protein binding interface to mediate protein-protein interactions. In skeletal muscle, FHL1 is involved in sarcomere assembly, differentiation, growth, and biomechanical stress. Muscle abnormalities may play a major role in congenital clubfoot (CCF) deformity during fetal development. Thus, identifying the interactions of FHL1 could provide important new insights into its functional role in both skeletal muscle development and CCF pathogenesis. Using proteins derived from rat L6GNR4 myoblastocytes, we detected FHL1 interacting proteins by immunoprecipitation. Samples were analyzed by liquid chromatography mass spectrometry (LC-MS). Dynamic gene expression of FHL1 was studied. Additionally, the expression of the possible interacting proteins gamma-actin and non-muscle myosin IIB, which were isolated from the lower limbs of E14, E15, E17, E18, E20 rat embryos or from adult skeletal muscle was analyzed. Potential interacting proteins isolated from E17 lower limbs were verified by immunoprecipitation, and co-localization in adult gastrocnemius muscle was visualized by fluorescence microscopy. FHL1 expression was associated with skeletal muscle differentiation. E17 was found to be the critical time-point for skeletal muscle differentiation in the lower limbs of rat embryos. We also identified gamma-actin and non-muscle myosin IIB as potential binding partners of FHL1, and both were expressed in adult skeletal muscle. We then demonstrated that FHL1 exists as part of a complex, which binds gamma-actin and non-muscle myosin IIB.
Collapse
Affiliation(s)
- Lili Wang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
- * E-mail:
| | - Jianing Miao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Lianyong Li
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Di Wu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yi Zhang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Zhaohong Peng
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Lijun Zhang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Kailai Sun
- Department of Medical Genetics, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Alp/Enigma family proteins cooperate in Z-disc formation and myofibril assembly. PLoS Genet 2013; 9:e1003342. [PMID: 23505387 PMCID: PMC3591300 DOI: 10.1371/journal.pgen.1003342] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/10/2013] [Indexed: 11/19/2022] Open
Abstract
The Drosophila Alp/Enigma family protein Zasp52 localizes to myotendinous junctions and Z-discs. It is required for terminal muscle differentiation and muscle attachment. Its vertebrate ortholog ZASP/Cypher also localizes to Z-discs, interacts with α-actinin through its PDZ domain, and is involved in Z-disc maintenance. Human mutations in ZASP cause myopathies and cardiomyopathies. Here we show that Drosophila Zasp52 is one of the earliest markers of Z-disc assembly, and we use a Zasp52-GFP fusion to document myofibril assembly by live imaging. We demonstrate that Zasp52 is required for adult Z-disc stability and pupal myofibril assembly. In addition, we show that two closely related proteins, Zasp66 and the newly identified Zasp67, are also required for adult Z-disc stability and are participating with Zasp52 in Z-disc assembly resulting in more severe, synergistic myofibril defects in double mutants. Zasp52 and Zasp66 directly bind to α-actinin, and they can also form a ternary complex. Our results indicate that Alp/Enigma family members cooperate in Z-disc assembly and myofibril formation; and we propose, based on sequence analysis, a novel class of PDZ domain likely involved in α-actinin binding.
Collapse
|
21
|
Recent advances in cardiovascular proteomics. J Proteomics 2012; 81:3-14. [PMID: 23153792 DOI: 10.1016/j.jprot.2012.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 10/10/2012] [Accepted: 10/31/2012] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVDs) are the major source of global morbidity and death and more people die annually from CVDs than from any other cause. These diseases can occur quickly, as seen in acute myocardial infarction (AMI), or progress slowly over years as with chronic heart failure. Advances in mass spectrometry detection and analysis, together with improved isolation and enrichment techniques allowing for the separation of organelles and membrane proteins, now allow for the indepth analysis of the cardiac proteome. Here we outline current insights that have been provided through cardiovascular proteomics, and discuss studies that have developed innovative technologies which permit the examination of the protein complement in specific organelles including exosomes and secreted proteins. We highlight these foundational studies and illustrate how they are providing the technologies and tools which are now being applied to further study cardiovascular disease; provide new diagnostic markers and potentially new methods of cardiac patient management with identification of novel drug targets. This article is part of a Special Issue entitled: From protein structures to clinical applications.
Collapse
|
22
|
Chis R, Sharma P, Bousette N, Miyake T, Wilson A, Backx PH, Gramolini AO. α-Crystallin B prevents apoptosis after H2O2 exposure in mouse neonatal cardiomyocytes. Am J Physiol Heart Circ Physiol 2012; 303:H967-78. [PMID: 22904156 PMCID: PMC3706333 DOI: 10.1152/ajpheart.00040.2012] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α-Crystallin B (cryAB) is the most abundant small heat shock protein in cardiomyocytes (CMs) and has been shown to have potent antiapoptotic properties. Because the mechanism by which cryAB prevents apoptosis has not been fully characterized, we examined its protective effects at the cellular level by silencing cryAB in mouse neonatal CMs using lentivector-mediated transduction of short hairpin RNAs. Subcellular fractionation of whole hearts showed that cryAB is cytosolic under control conditions, and after H(2)O(2) exposure, it translocates to the mitochondria. Phosphorylated cryAB (PcryAB) is mainly associated with the mitochondria, and any residual cytosolic PcryAB translocates to the mitochondria after H(2)O(2) exposure. H(2)O(2) exposure caused increases in cryAB and PcryAB levels, and cryAB silencing resulted in increased levels of apoptosis after exposure to H(2)O(2). Coimmunoprecipitation assays revealed an apparent interaction of both cryAB and PcryAB with mitochondrial voltage-dependent anion channels (VDAC), translocase of outer mitochondrial membranes 20 kDa (TOM 20), caspase 3, and caspase 12 in mouse cardiac tissue. Our results are consistent with the conclusion that the cardioprotective effects of cryAB are mediated by its translocation from the cytosol to the mitochondria under conditions of oxidative stress and that cryAB interactions with VDAC, TOM 20, caspase 3, and caspase 12 may be part of its protective mechanism.
Collapse
Affiliation(s)
- Roxana Chis
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5G 1L6
| | | | | | | | | | | | | |
Collapse
|
23
|
Li A, Ponten F, dos Remedios CG. The interactome of LIM domain proteins: The contributions of LIM domain proteins to heart failure and heart development. Proteomics 2012; 12:203-25. [DOI: 10.1002/pmic.201100492] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 12/22/2022]
|
24
|
Abstract
During the past 2 years, considerable progress in the field of four and a half LIM domain protein 1 (FHL1)-related myopathies has led to the identification of a growing number of FHL1 mutations. This genetic progress has uncovered crucial pathophysiological concepts, thus redefining clinical phenotypes. Important new characterizations include 4 distinct human myopathies: reducing body myopathy, X-linked myopathy with postural muscle atrophy, Emery-Dreifuss muscular dystrophy, and scapuloperoneal myopathy. Additionally, FHL1 mutations have been discovered in rigid spine syndrome and in a single family with contractures, rigid spine, and cardiomyopathy. In this review, we focus on the clinical phenotypes, which we correlate with the novel genetic and histological findings encountered within FHL1-related myopathies. This correlation will frequently lead to a considerably expanded clinical spectrum associated with a given FHL1 mutation.
Collapse
|
25
|
Four and a half LIM protein 1C (FHL1C): a binding partner for voltage-gated potassium channel K(v1.5). PLoS One 2011; 6:e26524. [PMID: 22053194 PMCID: PMC3203871 DOI: 10.1371/journal.pone.0026524] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 09/27/2011] [Indexed: 12/19/2022] Open
Abstract
Four-and-a-half LIM domain protein 1 isoform A (FHL1A) is predominantly expressed in skeletal and cardiac muscle. Mutations in the FHL1 gene are causative for several types of hereditary myopathies including X-linked myopathy with postural muscle atrophy (XMPMA). We here studied myoblasts from XMPMA patients. We found that functional FHL1A protein is completely absent in patient myoblasts. In parallel, expression of FHL1C is either unaffected or increased. Furthermore, a decreased proliferation rate of XMPMA myoblasts compared to controls was observed but an increased number of XMPMA myoblasts was found in the G0/G1 phase. Furthermore, low expression of Kv1.5, a voltage-gated potassium channel known to alter myoblast proliferation during the G1 phase and to control repolarization of action potential, was detected. In order to substantiate a possible relation between Kv1.5 and FHL1C, a pull-down assay was performed. A physical and direct interaction of both proteins was observed in vitro. In addition, confocal microscopy revealed substantial colocalization of FHL1C and Kv1.5 within atrial cells, supporting a possible interaction between both proteins in vivo. Two-electrode voltage clamp experiments demonstrated that coexpression of Kv1.5 with FHL1C in Xenopus laevis oocytes markedly reduced K+ currents when compared to oocytes expressing Kv1.5 only. We here present the first evidence on a biological relevance of FHL1C.
Collapse
|
26
|
Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, Elefanty AG, Gramolini A, Keller G. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nat Biotechnol 2011; 29:1011-8. [PMID: 22020386 DOI: 10.1038/nbt.2005] [Citation(s) in RCA: 393] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 09/19/2011] [Indexed: 11/09/2022]
Abstract
To identify cell-surface markers specific to human cardiomyocytes, we screened cardiovascular cell populations derived from human embryonic stem cells (hESCs) against a panel of 370 known CD antibodies. This screen identified the signal-regulatory protein alpha (SIRPA) as a marker expressed specifically on cardiomyocytes derived from hESCs and human induced pluripotent stem cells (hiPSCs), and PECAM, THY1, PDGFRB and ITGA1 as markers of the nonmyocyte population. Cell sorting with an antibody against SIRPA allowed for the enrichment of cardiac precursors and cardiomyocytes from hESC/hiPSC differentiation cultures, yielding populations of up to 98% cardiac troponin T-positive cells. When plated in culture, SIRPA-positive cells were contracting and could be maintained over extended periods of time. These findings provide a simple method for isolating populations of cardiomyocytes from human pluripotent stem cell cultures, and thereby establish a readily adaptable technology for generating large numbers of enriched cardiomyocytes for therapeutic applications.
Collapse
Affiliation(s)
- Nicole C Dubois
- McEwen Centre for Regenerative Medicine, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|