1
|
Liu Y, Jiang Y, Ma T, Dong W, Yang P, Peng L, Wang B, Wu C, Li Z, Zhang H, Sun Y, Niu Y, Ding Y. Cardiomyocyte-specific activation of the sarcomere-localized Dnajb6b chaperone causes cardiomyopathy and heart failure through upregulated sarcoplasmic reticulum stress. Life Sci 2025; 374:123711. [PMID: 40360088 DOI: 10.1016/j.lfs.2025.123711] [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: 02/09/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
AIMS Despite abundant expression of DNAJB6 gene in the heart, its roles in cardiac diseases remain underexplored. We aimed to investigate the function of its zebrafish (Danio rerio) ortholog, the dnajb6b gene, in cardiomyopathy and heart failure. MATERIALS AND METHODS Both loss-of-function mutation and gain-of-function transgenic approaches were employed in zebrafish. High frequency echocardiography was performed to evaluate cardiac function indices in adult zebrafish. 4-phenylbutyric acid (4-PBA) was used to pharmacologically inhibit sarcoplasmic reticulum (SR) stress in zebrafish. Western blot was carried out to determine expression of DNAJB6 isoforms in human patients' heart tissues. KEY FINDINGS Global loss-of-function mutations affecting both the sarcomere-localized short (Dnajb6b[S]) and nucleus-localized long (Dnajb6b[L]) isoforms appeared phenotypically normal. In contrast, cardiomyocyte-specific overexpression of a truncated, sarcomere-localized Dnajb6b(L) isoform (Dnajb6b[∆L]) led to severe cardiomyopathy and heart failure phenotypes. Mechanistically, Dnajb6b responded to sarcoplasmic reticulum (SR) stress and activation of Dnajb6b(∆L) resulted in elevated SR stress, accumulation of ubiquitinated protein aggregation, and aberrant activation of autophagy. 4-PBA treatment partially rescued cardiac dysfunction and extended the lifespan of zebrafish with cardiomyocyte-specific activation of Dnajb6b(∆L). Finally, elevated expression of both DNAJB6(S) and DNAJB6(L) isoforms was detected in failing human hearts, supporting their clinical relevance. SIGNIFICANCE Gain-of-function mutation in Dnajb6b(∆L) isoform causes cardiomyopathy and heart failure, likely mediated by elevated SR stress. This study enhances our understanding of Dnajb6's role in cardiac proteostasis and highlights its potential as a therapeutic target for the treatment of cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Yuting Liu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yajie Jiang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Taiwei Ma
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Wenjing Dong
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Peng Yang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lixia Peng
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Baokun Wang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Hong Zhang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yuanchao Sun
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yujuan Niu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yonghe Ding
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
2
|
Angom RS, Singh M, Muhammad H, Varanasi SM, Mukhopadhyay D. Zebrafish as a Versatile Model for Cardiovascular Research: Peering into the Heart of the Matter. Cells 2025; 14:531. [PMID: 40214485 PMCID: PMC11988917 DOI: 10.3390/cells14070531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. A total of 17.5 million people died of CVDs in the year 2012, accounting for 31% of all deaths globally. Vertebrate animal models have been used to understand cardiac disease biology, as the cellular, molecular, and physiological aspects of human CVDs can be replicated closely in these organisms. Zebrafish is a popular model organism offering an arsenal of genetic tools that allow the rapid in vivo analysis of vertebrate gene function and disease conditions. It has a short breeding cycle, high fecundity, optically transparent embryos, rapid internal organ development, and easy maintenance. This review aims to give readers an overview of zebrafish cardiac biology and a detailed account of heart development in zebrafish and its comparison with humans and the conserved genetic circuitry. We also discuss the contributions made in CVD research using the zebrafish model. The first part of this review focuses on detailed information on the morphogenetic and differentiation processes in early cardiac development. The overlap and divergence of the human heart's genetic circuitry, structure, and physiology are emphasized wherever applicable. In the second part of the review, we overview the molecular tools and techniques available to dissect gene function and expression in zebrafish, with special mention of the use of these tools in cardiac biology.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Meghna Singh
- Department of Pathology and Lab Medicine, University of California, Los Angeles, CA 92093, USA;
| | - Huzaifa Muhammad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| |
Collapse
|
3
|
Ding Y, Wang M, Bu H, Li J, Lin X, Xu X. Application of an F0-based genetic assay in adult zebrafish to identify modifier genes of an inherited cardiomyopathy. Dis Model Mech 2023; 16:dmm049427. [PMID: 35481478 PMCID: PMC9239171 DOI: 10.1242/dmm.049427] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 01/08/2023] Open
Abstract
Modifier genes contribute significantly to our understanding of pathophysiology in human diseases; however, effective approaches to identify modifier genes are still lacking. Here, we aim to develop a rapid F0-based genetic assay in adult zebrafish using the bag3 gene knockout (bag3e2/e2) cardiomyopathy model as a paradigm. First, by utilizing a classic genetic breeding approach, we identified dnajb6b as a deleterious modifier gene for bag3 cardiomyopathy. Next, we established an F0-based genetic assay in adult zebrafish through injection of predicted microhomology-mediated end joining (MMEJ)-inducing single guide RNA/Cas9 protein complex. We showed that effective gene knockdown is maintained in F0 adult fish, enabling recapitulation of both salutary modifying effects of the mtor haploinsufficiency and deleterious modifying effects of the dnajb6b gene on bag3 cardiomyopathy. We finally deployed the F0-based genetic assay to screen differentially expressed genes in the bag3 cardiomyopathy model. As a result, myh9b was identified as a novel modifier gene for bag3 cardiomyopathy. Together, these data prove the feasibility of an F0 adult zebrafish-based genetic assay that can be effectively used to discover modifier genes for inherited cardiomyopathy.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mingmin Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiothoracic Surgery, Xiangfan Hospital, Central South University, Changsha 410008, China
| | - Jiarong Li
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Surgery, The Second Xiangfan Hospital of Central South University, Changsha 410011, China
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Li X, Li M. The application of zebrafish patient-derived xenograft tumor models in the development of antitumor agents. Med Res Rev 2023; 43:212-236. [PMID: 36029178 DOI: 10.1002/med.21924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/09/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high-throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient-derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting-edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
5
|
Ding Y, Lang D, Yan J, Bu H, Li H, Jiao K, Yang J, Ni H, Morotti S, Le T, Clark KJ, Port J, Ekker SC, Cao H, Zhang Y, Wang J, Grandi E, Li Z, Shi Y, Li Y, Glukhov AV, Xu X. A phenotype-based forward genetic screen identifies Dnajb6 as a sick sinus syndrome gene. eLife 2022; 11:e77327. [PMID: 36255053 PMCID: PMC9642998 DOI: 10.7554/elife.77327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Previously we showed the generation of a protein trap library made with the gene-break transposon (GBT) in zebrafish (Danio rerio) that could be used to facilitate novel functional genome annotation towards understanding molecular underpinnings of human diseases (Ichino et al, 2020). Here, we report a significant application of this library for discovering essential genes for heart rhythm disorders such as sick sinus syndrome (SSS). SSS is a group of heart rhythm disorders caused by malfunction of the sinus node, the heart's primary pacemaker. Partially owing to its aging-associated phenotypic manifestation and low expressivity, molecular mechanisms of SSS remain difficult to decipher. From 609 GBT lines screened, we generated a collection of 35 zebrafish insertional cardiac (ZIC) mutants in which each mutant traps a gene with cardiac expression. We further employed electrocardiographic measurements to screen these 35 ZIC lines and identified three GBT mutants with SSS-like phenotypes. More detailed functional studies on one of the arrhythmogenic mutants, GBT411, in both zebrafish and mouse models unveiled Dnajb6 as a novel SSS causative gene with a unique expression pattern within the subpopulation of sinus node pacemaker cells that partially overlaps with the expression of hyperpolarization activated cyclic nucleotide gated channel 4 (HCN4), supporting heterogeneity of the cardiac pacemaker cells.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Di Lang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
- Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Jianhua Yan
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Cardiothoracic Surgery, Xiangya Hospital, Central South UniversityChangshaChina
| | - Hongsong Li
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Cardiovascular Medicine, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health ScienceShanghaiChina
| | - Kunli Jiao
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Jingchun Yang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Haibo Ni
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Stefano Morotti
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Tai Le
- Department of Biomedical Engineering, University of California, IrvineIrvineUnited States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Jenna Port
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Hung Cao
- Department of Biomedical Engineering, University of California, IrvineIrvineUnited States
- Department of Electrical Engineering and Computer Science, University of California, IrvineIrvineUnited States
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland School of MedicineBaltimoreUnited States
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Eleonora Grandi
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Yigang Li
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Alexey V Glukhov
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| |
Collapse
|
6
|
Abstract
Heart disease is the leading cause of death worldwide. Despite decades of research, most heart pathologies have limited treatments, and often the only curative approach is heart transplantation. Thus, there is an urgent need to develop new therapeutic approaches for treating cardiac diseases. Animal models that reproduce the human pathophysiology are essential to uncovering the biology of diseases and discovering therapies. Traditionally, mammals have been used as models of cardiac disease, but the cost of generating and maintaining new models is exorbitant, and the studies have very low throughput. In the last decade, the zebrafish has emerged as a tractable model for cardiac diseases, owing to several characteristics that made this animal popular among developmental biologists. Zebrafish fertilization and development are external; embryos can be obtained in high numbers, are cheap and easy to maintain, and can be manipulated to create new genetic models. Moreover, zebrafish exhibit an exceptional ability to regenerate their heart after injury. This review summarizes 25 years of research using the zebrafish to study the heart, from the classical forward screenings to the contemporary methods to model mutations found in patients with cardiac disease. We discuss the advantages and limitations of this model organism and introduce the experimental approaches exploited in zebrafish, including forward and reverse genetics and chemical screenings. Last, we review the models used to induce cardiac injury and essential ideas derived from studying natural regeneration. Studies using zebrafish have the potential to accelerate the discovery of new strategies to treat cardiac diseases.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA
| |
Collapse
|
7
|
Karamanos TK, Clore GM. Large Chaperone Complexes Through the Lens of Nuclear Magnetic Resonance Spectroscopy. Annu Rev Biophys 2022; 51:223-246. [PMID: 35044800 DOI: 10.1146/annurev-biophys-090921-120150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular chaperones are the guardians of the proteome inside the cell. Chaperones recognize and bind unfolded or misfolded substrates, thereby preventing further aggregation; promoting correct protein folding; and, in some instances, even disaggregating already formed aggregates. Chaperones perform their function by means of an array of weak protein-protein interactions that take place over a wide range of timescales and are therefore invisible to structural techniques dependent upon the availability of highly homogeneous samples. Nuclear magnetic resonance (NMR) spectroscopy, however, is ideally suited to study dynamic, rapidly interconverting conformational states and protein-protein interactions in solution, even if these involve a high-molecular-weight component. In this review, we give a brief overview of the principles used by chaperones to bind their client proteins and describe NMR methods that have emerged as valuable tools to probe chaperone-substrate and chaperone-chaperone interactions. We then focus on a few systems for which the application of these methods has greatly increased our understanding of the mechanisms underlying chaperone functions. Expected final online publication date for the Annual Review of Biophysics, Volume 51 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Theodoros K Karamanos
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom;
| | - G Marius Clore
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
8
|
Bu H, Ding Y, Li J, Zhu P, Shih YH, Wang M, Zhang Y, Lin X, Xu X. Inhibition of mTOR or MAPK ameliorates vmhcl/myh7 cardiomyopathy in zebrafish. JCI Insight 2021; 6:154215. [PMID: 34935644 PMCID: PMC8783688 DOI: 10.1172/jci.insight.154215] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/03/2021] [Indexed: 01/25/2023] Open
Abstract
Myosin heavy chain 7 (MYH7) is a major causative gene for hypertrophic cardiomyopathy, but the affected signaling pathways and therapeutics remain elusive. In this research, we identified ventricle myosin heavy chain like (vmhcl) as a zebrafish homolog of human MYH7, and we generated vmhcl frameshift mutants. We noted vmhcl-based embryonic cardiac dysfunction (VEC) in the vmhcl homozygous mutants and vmhcl-based adult cardiomyopathy (VAC) phenotypes in the vmhcl heterozygous mutants. Using the VEC model, we assessed 7 known cardiomyopathy signaling pathways pharmacologically and 11 candidate genes genetically via CRISPR/Cas9 genome editing technology based on microhomology-mediated end joining (MMEJ). Both studies converged on therapeutic benefits of mTOR or mitogen-activated protein kinase (MAPK) inhibition of VEC. While mTOR inhibition rescued the enlarged nuclear size of cardiomyocytes, MAPK inhibition restored the prolonged cell shape in the VEC model. The therapeutic effects of mTOR and MAPK inhibition were later validated in the VAC model. Together, vmhcl/myh7 loss of function is sufficient to induce cardiomyopathy in zebrafish. The VEC and VAC models in zebrafish are amenable to both efficient genetic and chemical genetic tools, offering a rapid in vivo platform for discovering candidate signaling pathways of MYH7 cardiomyopathy.
Collapse
Affiliation(s)
- Haisong Bu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiothoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jiarong Li
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yu-Huan Shih
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mingmin Wang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
In vivo identification and validation of novel potential predictors for human cardiovascular diseases. PLoS One 2021; 16:e0261572. [PMID: 34919578 PMCID: PMC8682894 DOI: 10.1371/journal.pone.0261572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022] Open
Abstract
Genetics crucially contributes to cardiovascular diseases (CVDs), the global leading cause of death. Since the majority of CVDs can be prevented by early intervention there is a high demand for the identification of predictive causative genes. While genome wide association studies (GWAS) correlate genes and CVDs after diagnosis and provide a valuable resource for such causative candidate genes, often preferentially those with previously known or suspected function are addressed further. To tackle the unaddressed blind spot of understudied genes, we particularly focused on the validation of human heart phenotype-associated GWAS candidates with little or no apparent connection to cardiac function. Building on the conservation of basic heart function and underlying genetics from fish to human we combined CRISPR/Cas9 genome editing of the orthologs of human GWAS candidates in isogenic medaka with automated high-throughput heart rate analysis. Our functional analyses of understudied human candidates uncovered a prominent fraction of heart rate associated genes from adult human patients impacting on the heart rate in embryonic medaka already in the injected generation. Following this pipeline, we identified 16 GWAS candidates with potential diagnostic and predictive power for human CVDs.
Collapse
|
10
|
Bensimon-Brito A, Boezio GLM, Cardeira-da-Silva J, Wietelmann A, Ramkumar S, Lundegaard PR, Helker CSM, Ramadass R, Piesker J, Nauerth A, Mueller C, Stainier DYR. Integration of multiple imaging platforms to uncover cardiovascular defects in adult zebrafish. Cardiovasc Res 2021; 118:2665-2687. [PMID: 34609500 PMCID: PMC9491864 DOI: 10.1093/cvr/cvab310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
Aims Mammalian models have been instrumental in investigating adult heart function and human disease. However, electrophysiological differences with human hearts and high costs motivate the need for non-mammalian models. The zebrafish is a well-established genetic model to study cardiovascular development and function; however, analysis of cardiovascular phenotypes in adult specimens is particularly challenging as they are opaque. Methods and results Here, we optimized and combined multiple imaging techniques including echocardiography, magnetic resonance imaging, and micro-computed tomography to identify and analyse cardiovascular phenotypes in adult zebrafish. Using alk5a/tgfbr1a mutants as a case study, we observed morphological and functional cardiovascular defects that were undetected with conventional approaches. Correlation analysis of multiple parameters revealed an association between haemodynamic defects and structural alterations of the heart, as observed clinically. Conclusion We report a new, comprehensive, and sensitive platform to identify otherwise indiscernible cardiovascular phenotypes in adult zebrafish.
Collapse
Affiliation(s)
- Anabela Bensimon-Brito
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Giulia L M Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
| | - João Cardeira-da-Silva
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Scientific Service Group MRI and µ-CT, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Srinath Ramkumar
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Pia R Lundegaard
- Laboratory for Molecular Cardiology, Department of Cardiology, Vascular, Pulmonary and Infectious Diseases, University Hospital of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Radhan Ramadass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
| |
Collapse
|
11
|
Disruption of MAP7D1 Gene Function Increases the Risk of Doxorubicin-Induced Cardiomyopathy and Heart Failure. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8569921. [PMID: 34327238 PMCID: PMC8302367 DOI: 10.1155/2021/8569921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022]
Abstract
Doxorubicin is a cornerstone chemotherapeutic drug widely used to treat various cancers; its dose-dependent cardiomyopathy, however, is one of the leading causes of treatment-associated mortality in cancer survivors. Patients' threshold doses leading to doxorubicin-induced cardiomyopathy (DIC) and heart failure are highly variable, mostly due to genetic variations in individuals' genomes. However, genetic susceptibility to DIC remains largely unidentified. Here, we combined a genetic approach in the zebrafish (Danio rerio) animal model with a genome-wide association study (GWAS) in humans to identify genetic susceptibility to DIC and heart failure. We firstly reported the cardiac and skeletal muscle-specific expression and sarcomeric localization of the microtubule-associated protein 7 domain-containing protein 1b (Map7d1b) in zebrafish, followed by expression validation in mice. We then revealed that disruption of the map7d1b gene function exaggerated DIC effects in adult zebrafish. Mechanistically, the exacerbated DIC are likely conveyed by impaired autophagic degradation and elevated protein aggregation. Lastly, we identified 2 MAP7D1 gene variants associated with cardiac functional decline and heart failure in cancer patients who received doxorubicin therapy. Together, this study identifies MAP7D1 as a clinically relevant susceptibility gene to DIC and heart failure, providing useful information to stratify cancer patients with a high risk of incurring severe cardiomyopathy and heart failure after receiving chemotherapy.
Collapse
|
12
|
Kim M, Lu L, Dvornikov AV, Ma X, Ding Y, Zhu P, Olson TM, Lin X, Xu X. TFEB Overexpression, Not mTOR Inhibition, Ameliorates RagC S75Y Cardiomyopathy. Int J Mol Sci 2021; 22:5494. [PMID: 34071043 PMCID: PMC8197163 DOI: 10.3390/ijms22115494] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
A de novo missense variant in Rag GTPase protein C (RagCS75Y) was recently identified in a syndromic dilated cardiomyopathy (DCM) patient. However, its pathogenicity and the related therapeutic strategy remain unclear. We generated a zebrafish RragcS56Y (corresponding to human RagCS75Y) knock-in (KI) line via TALEN technology. The KI fish manifested cardiomyopathy-like phenotypes and poor survival. Overexpression of RagCS75Y via adenovirus infection also led to increased cell size and fetal gene reprogramming in neonatal rat ventricle cardiomyocytes (NRVCMs), indicating a conserved mechanism. Further characterization identified aberrant mammalian target of rapamycin complex 1 (mTORC1) and transcription factor EB (TFEB) signaling, as well as metabolic abnormalities including dysregulated autophagy. However, mTOR inhibition failed to ameliorate cardiac phenotypes in the RagCS75Y cardiomyopathy models, concomitant with a failure to promote TFEB nuclear translocation. This observation was at least partially explained by increased and mTOR-independent physical interaction between RagCS75Y and TFEB in the cytosol. Importantly, TFEB overexpression resulted in more nuclear TFEB and rescued cardiomyopathy phenotypes. These findings suggest that S75Y is a pathogenic gain-of-function mutation in RagC that leads to cardiomyopathy. A primary pathological step of RagCS75Y cardiomyopathy is defective mTOR-TFEB signaling, which can be corrected by TFEB overexpression, but not mTOR inhibition.
Collapse
Affiliation(s)
- Maengjo Kim
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| | - Linghui Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Alexey V. Dvornikov
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| | - Timothy M. Olson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN 55901, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA; (M.K.); (L.L.); (A.V.D.); (X.M.); (Y.D.); (P.Z.); (X.L.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55901, USA;
| |
Collapse
|
13
|
Rare Pathogenic Variants in Mitochondrial and Inflammation-Associated Genes May Lead to Inflammatory Cardiomyopathy in Chagas Disease. J Clin Immunol 2021; 41:1048-1063. [PMID: 33660144 PMCID: PMC8249271 DOI: 10.1007/s10875-021-01000-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/15/2021] [Indexed: 01/21/2023]
Abstract
Abstract Cardiomyopathies are an important cause of heart failure and sudden cardiac death. Little is known about the role of rare genetic variants in inflammatory cardiomyopathy. Chronic Chagas disease cardiomyopathy (CCC) is an inflammatory cardiomyopathy prevalent in Latin America, developing in 30% of the 6 million patients chronically infected by the protozoan Trypanosoma cruzi, while 60% remain free of heart disease (asymptomatic (ASY)). The cytokine interferon-γ and mitochondrial dysfunction are known to play a major pathogenetic role. Chagas disease provides a unique model to probe for genetic variants involved in inflammatory cardiomyopathy. Methods We used whole exome sequencing to study nuclear families containing multiple cases of Chagas disease. We searched for rare pathogenic variants shared by all family members with CCC but absent in infected ASY siblings and in unrelated ASY. Results We identified heterozygous, pathogenic variants linked to CCC in all tested families on 22 distinct genes, from which 20 were mitochondrial or inflammation-related – most of the latter involved in proinflammatory cytokine production. Significantly, incubation with IFN-γ on a human cardiomyocyte line treated with an inhibitor of dihydroorotate dehydrogenase brequinar (enzyme showing a loss-of-function variant in one family) markedly reduced mitochondrial membrane potential (ΔψM), indicating mitochondrial dysfunction. Conclusion Mitochondrial dysfunction and inflammation may be genetically determined in CCC, driven by rare genetic variants. We hypothesize that CCC-linked genetic variants increase mitochondrial susceptibility to IFN-γ-induced damage in the myocardium, leading to the cardiomyopathy phenotype in Chagas disease. This mechanism may also be operative in other inflammatory cardiomyopathies. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-021-01000-y.
Collapse
|
14
|
Ding Y, Bu H, Xu X. Modeling Inherited Cardiomyopathies in Adult Zebrafish for Precision Medicine. Front Physiol 2020; 11:599244. [PMID: 33329049 PMCID: PMC7717946 DOI: 10.3389/fphys.2020.599244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiomyopathies are a highly heterogeneous group of heart muscle disorders. More than 100 causative genes have been linked to various cardiomyopathies, which explain about half of familial cardiomyopathy cases. More than a dozen candidate therapeutic signaling pathways have been identified; however, precision medicine is not being used to treat the various types of cardiomyopathy because knowledge is lacking for how to tailor treatment plans for different genetic causes. Adult zebrafish (Danio rerio) have a higher throughout than rodents and are an emerging vertebrate model for studying cardiomyopathy. Herein, we review progress in the past decade that has proven the feasibility of this simple vertebrate for modeling inherited cardiomyopathies of distinct etiology, identifying effective therapeutic strategies for a particular type of cardiomyopathy, and discovering new cardiomyopathy genes or new therapeutic strategies via a forward genetic approach. On the basis of this progress, we discuss future research that would benefit from integrating this emerging model, including discovery of remaining causative genes and development of genotype-based therapies. Studies using this efficient vertebrate model are anticipated to significantly accelerate the implementation of precision medicine for inherited cardiomyopathies.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States.,Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
15
|
An S/T motif controls reversible oligomerization of the Hsp40 chaperone DNAJB6b through subtle reorganization of a β sheet backbone. Proc Natl Acad Sci U S A 2020; 117:30441-30450. [PMID: 33199640 DOI: 10.1073/pnas.2020306117] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chaperone oligomerization is often a key aspect of their function. Irrespective of whether chaperone oligomers act as reservoirs for active monomers or exhibit a chaperoning function themselves, understanding the mechanism of oligomerization will further our understanding of how chaperones maintain the proteome. Here, we focus on the class-II Hsp40, human DNAJB6b, a highly efficient inhibitor of protein self-assembly in vivo and in vitro that forms functional oligomers. Using single-quantum methyl-based relaxation dispersion NMR methods we identify critical residues for DNAJB6b oligomerization in its C-terminal domain (CTD). Detailed solution NMR studies on the structure of the CTD showed that a serine/threonine-rich stretch causes a backbone twist in the N-terminal β strand, stabilizing the monomeric form. Quantitative analysis of an array of NMR relaxation-based experiments (including Carr-Purcell-Meiboom-Gill relaxation dispersion, off-resonance R 1ρ profiles, lifetime line broadening, and exchange-induced shifts) on the CTD of both wild type and a point mutant (T142A) within the S/T region of the first β strand delineates the kinetics of the interconversion between the major twisted-monomeric conformation and a more regular β strand configuration in an excited-state dimer, as well as exchange of both monomer and dimer species with high-molecular-weight oligomers. These data provide insights into the molecular origins of DNAJB6b oligomerization. Further, the results reported here have implications for the design of β sheet proteins with tunable self-assembling properties and pave the way to an atomic-level understanding of amyloid inhibition.
Collapse
|
16
|
Ichino N, Serres MR, Urban RM, Urban MD, Treichel AJ, Schaefbauer KJ, Greif LE, Varshney GK, Skuster KJ, McNulty MS, Daby CL, Wang Y, Liao HK, El-Rass S, Ding Y, Liu W, Anderson JL, Wishman MD, Sabharwal A, Schimmenti LA, Sivasubbu S, Balciunas D, Hammerschmidt M, Farber SA, Wen XY, Xu X, McGrail M, Essner JJ, Burgess SM, Clark KJ, Ekker SC. Building the vertebrate codex using the gene breaking protein trap library. eLife 2020; 9:54572. [PMID: 32779569 PMCID: PMC7486118 DOI: 10.7554/elife.54572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
One key bottleneck in understanding the human genome is the relative under-characterization of 90% of protein coding regions. We report a collection of 1200 transgenic zebrafish strains made with the gene-break transposon (GBT) protein trap to simultaneously report and reversibly knockdown the tagged genes. Protein trap-associated mRFP expression shows previously undocumented expression of 35% and 90% of cloned genes at 2 and 4 days post-fertilization, respectively. Further, investigated alleles regularly show 99% gene-specific mRNA knockdown. Homozygous GBT animals in ryr1b, fras1, tnnt2a, edar and hmcn1 phenocopied established mutants. 204 cloned lines trapped diverse proteins, including 64 orthologs of human disease-associated genes with 40 as potential new disease models. Severely reduced skeletal muscle Ca2+ transients in GBT ryr1b homozygous animals validated the ability to explore molecular mechanisms of genetic diseases. This GBT system facilitates novel functional genome annotation towards understanding cellular and molecular underpinnings of vertebrate biology and human disease.
Collapse
Affiliation(s)
- Noriko Ichino
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - MaKayla R Serres
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Rhianna M Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Mark D Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Anthony J Treichel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Kyle J Schaefbauer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lauren E Greif
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Gaurav K Varshney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States.,Functional & Chemical Genomics Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Kimberly J Skuster
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Melissa S McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Camden L Daby
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ying Wang
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Hsin-Kai Liao
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Weibin Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Mark D Wishman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ankit Sabharwal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lisa A Schimmenti
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Clinical Genomics, Mayo Clinic, Rochester, United States.,Department of Otorhinolaryngology, Mayo Clinic, Rochester, United States
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Darius Balciunas
- Department of Biology, Temple University, Philadelphia, United States
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Steven Arthur Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Maura McGrail
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| |
Collapse
|
17
|
Cell stemness is maintained upon concurrent expression of RB and the mitochondrial ribosomal protein S18-2. Proc Natl Acad Sci U S A 2020; 117:15673-15683. [PMID: 32571933 PMCID: PMC7355020 DOI: 10.1073/pnas.1922535117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stemness encompasses the capability of a cell for self-renewal and differentiation. The stem cell maintains a balance between proliferation, quiescence, and regeneration via interactions with the microenvironment. Previously, we showed that ectopic expression of the mitochondrial ribosomal protein S18-2 (MRPS18-2) led to immortalization of primary fibroblasts, accompanied by induction of an embryonic stem cell (ESC) phenotype. Moreover, we demonstrated interaction between S18-2 and the retinoblastoma-associated protein (RB) and hypothesized that the simultaneous expression of RB and S18-2 is essential for maintaining cell stemness. Here, we experimentally investigated the role of S18-2 in cell stemness and differentiation. Concurrent expression of RB and S18-2 resulted in immortalization of Rb1 -/- primary mouse embryonic fibroblasts and in aggressive tumor growth in severe combined immunodeficiency mice. These cells, which express both RB and S18-2 at high levels, exhibited the potential to differentiate into various lineages in vitro, including osteogenic, chondrogenic, and adipogenic lineages. Mechanistically, S18-2 formed a multimeric protein complex with prohibitin and the ring finger protein 2 (RNF2). This molecular complex increased the monoubiquitination of histone H2ALys119, a characteristic trait of ESCs, by enhanced E3-ligase activity of RNF2. Furthermore, we found enrichment of KLF4 at the S18-2 promoter region and that the S18-2 expression is positively correlated with KLF4 levels. Importantly, knockdown of S18-2 in zebrafish larvae led to embryonic lethality. Collectively, our findings suggest an important role for S18-2 in cell stemness and differentiation and potentially also in cancerogenesis.
Collapse
|
18
|
Chen YC, Hsiao CC, Chen TW, Wu CC, Chao TY, Leung SY, Eng HL, Lee CP, Wang TY, Lin MC. Whole Genome DNA Methylation Analysis of Active Pulmonary Tuberculosis Disease Identifies Novel Epigenotypes: PARP9/ miR-505/ RASGRP4/ GNG12 Gene Methylation and Clinical Phenotypes. Int J Mol Sci 2020; 21:ijms21093180. [PMID: 32365959 PMCID: PMC7246806 DOI: 10.3390/ijms21093180] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/30/2022] Open
Abstract
We hypothesized that DNA methylation patterns may contribute to the development of active pulmonary tuberculosis (TB). Illumina’s DNA methylation 450 K assay was used to identify differentially methylated loci (DML) in a discovery cohort of 12 active pulmonary TB patients and 6 healthy subjects (HS). DNA methylation levels were validated in an independent cohort of 64 TB patients and 24 HS. Microarray analysis identified 1028 DMLs in TB patients versus HS, and 3747 DMLs in TB patients after versus before anti-TB treatment, while autophagy was the most enriched signaling pathway. In the validation cohort, PARP9 and miR505 genes were hypomethylated in the TB patients versus HS, while RASGRP4 and GNG12 genes were hypermethylated, with the former two further hypomethylated in those with delayed sputum conversion, systemic symptoms, or far advanced lesions. MRPS18B and RPTOR genes were hypomethylated in TB patients with pleural involvement. RASGRP4 gene hypermethylation and RPTOR gene down-regulation were associated with high mycobacterial burden. TB patients with WIPI2/GNG12 hypermethylation or MRPS18B/FOXO3 hypomethylation had lower one-year survival. In vitro ESAT6 and CFP10 stimuli of THP-1 cells resulted in DNA de-methylation changes of the PARP9, RASGRP4, WIPI2, and FOXO3 genes. In conclusions, aberrant DNA methylation over the PARP9/miR505/RASGRP4/GNG12 genes may contribute to the development of active pulmonary TB disease and its clinical phenotypes, while aberrant DNA methylation over the WIPI2/GNG12/MARPS18B/FOXO3 genes may constitute a determinant of long-term outcomes.
Collapse
Affiliation(s)
- Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
- Graduate Institute of Clinical Medical Sciences and Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (Y.-C.C.); (M.-C.L.); Tel.: +886-975056041 (Y.-C.C.); +886-7-731-7123 (ext 8199) (M.-C.L.)
| | - Chang-Chun Hsiao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
- Graduate Institute of Clinical Medical Sciences and Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ting-Wen Chen
- Molecular Medicine Research Center, and Bioinformatics Center, Chang Gung University, Taoyuan 33302, Taiwan;
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30068, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chao-Chien Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
| | - Tung-Ying Chao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
| | - Sum-Yee Leung
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
| | - Hock-Liew Eng
- Division of Clinical Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Chiu-Ping Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
| | - Ting-Ya Wang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-C.H.); (C.-C.W.); (T.-Y.C.); (S.-Y.L.); (C.-P.L.); (T.-Y.W.)
- Correspondence: (Y.-C.C.); (M.-C.L.); Tel.: +886-975056041 (Y.-C.C.); +886-7-731-7123 (ext 8199) (M.-C.L.)
| |
Collapse
|
19
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|
20
|
Xie X, Jin Y, Ma Z, Tang S, Peng H, Giesy JP, Liu H. Underlying mechanisms of reproductive toxicity caused by multigenerational exposure of 2, bromo-4, 6-dinitroaniline (BDNA) to Zebrafish (Danio rerio) at environmental relevant levels. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 216:105285. [PMID: 31546070 DOI: 10.1016/j.aquatox.2019.105285] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 06/10/2023]
Abstract
2-bromo-4, 6-dinitroaniline (BDNA) is a mutagenic aromatic amine involved in the production and degradation of Disperse blue 79, one of the most extensively used brominated azo dyes. In our previous study, a multigenerational exposure of BDNA (0.5, 5, 50 and 500 μg/L) to zebrafish from F0 adult to F2 larvae including a recovery group in F2 larvae was conducted. The effects on apical points observed in individuals and the long-term effects predicted on population were all related to reproduction. In this study, we performed molecular analysis to elucidate the underlying mechanisms of the reproductive toxicity of BDNA. In F1 generation, measurement of vitellogenin and transcription levels of genes associated with hypothalamus-pituitary-gland (HPG) axis, estrogen receptor (ER) and androgen receptor (AR) were conducted. There was a decrease in VTG level in the blood of F1 female fish and transcription of genes related to ER was more affected than that of genes related to AR. These results were consistent with adverse effects that sexual differentiation was biased towards males and fecundity was impaired in a concentration-dependent manner in adults of F1 generation after 150 days exposure. In F2 generation, global gene transcriptions of F2 larvae were investigated. It was uncovered that processes related to apoptosis, development and DNA damage were strongly affected. Alterations to these biological pathways accounted for the irreversible parental influence on a significant decrease in hatchability and increase in abnormality of F2 larvae. All evidence suggested that the multigenerational exposure of BDNA posed lasting effects transmitted from parents to offspring that persisted after exposure ceased.
Collapse
Affiliation(s)
- Xianyi Xie
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Yaru Jin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Zhiyuan Ma
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Song Tang
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Hui Peng
- Department of Chemistry, University of Toronto, Ontario, M5S 3H6, Canada
| | - John P Giesy
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China; Department of Biomedical Veterinary Sciences and Toxicology Centre, University of Saskatchewan, Saskatoon, SKS7N 5B3, Canada
| | - Hongling Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
21
|
Le T, Lenning M, Clark I, Bhimani I, Fortunato J, Mash P, Xu X, Cao H. Acquisition, Processing and Analysis of Electrocardiogram in Awake Zebrafish. IEEE SENSORS JOURNAL 2019; 19:4283-4289. [PMID: 32855627 PMCID: PMC7449252 DOI: 10.1109/jsen.2019.2897789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Long-term monitoring of intrinsic electrocardiogram (ECG) in zebrafish plays a crucial role in heart disease studies as well as drug screening. In this work, we developed a polymer-based apparatus with embedded flexible thin-film electrodes to acquire ECG signals of awake zebrafish. The apparatus was made of polydimethylsiloxane (PDMS) using the molding technique with molds formed by 3D printing. A graphical user interface (GUI) was built in National Instruments LabView platform for real-time recording, processing and analysis. The program provided important features, such as signal de-noising, characteristic wave detection and anomaly detection. Further, it could operate on both real-time coming signals as well as previously-saved data, facilitating analysis and interpretation. We demonstrated the use of our system to investigate the effects of the anesthetic drug, namely Tricaine (MS-222), on cardiac electrophysiology of zebrafish, revealing promising findings. We speculate that our novel system may contribute to a host of studies in various disciplines using the zebrafish model.
Collapse
Affiliation(s)
- Tai Le
- HERO Laboratory, University of California Irvine, CA 92697, USA
| | | | - Isaac Clark
- HERO Laboratory, University of California Irvine, CA 92697, USA
| | | | | | - Paul Mash
- HERO Laboratory, University of California Irvine, CA 92697, USA
| | - Xiaolei Xu
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Hung Cao
- Director of the HERO Laboratory, Electrical Engineering and Computer Science Department, School of Engineering, University of California Irvine, CA 92697, USA
| |
Collapse
|
22
|
Dvornikov AV, de Tombe PP, Xu X. Phenotyping cardiomyopathy in adult zebrafish. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:116-125. [PMID: 29884423 PMCID: PMC6269218 DOI: 10.1016/j.pbiomolbio.2018.05.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/26/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is usually manifested by increased myofilament Ca2+ sensitivity, excessive contractility, and impaired relaxation. In contrast, dilated cardiomyopathy (DCM) originates from insufficient sarcomere contractility and reduced cardiac pump function, subsequently resulting in heart failure. The zebrafish has emerged as a new model of human cardiomyopathy with high-throughput screening, which will facilitate the discovery of novel genetic factors and the development of new therapies. Given the small hearts of zebrafish, better phenotyping tools are needed to discern different types of cardiomyopathy, such as HCM and DCM. This article reviews the existing models of cardiomyopathy, available morphologic and functional methods, and current understanding of the different types of cardiomyopathy in adult zebrafish.
Collapse
Affiliation(s)
- Alexey V Dvornikov
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Pieter P de Tombe
- University of Illinois at Chicago, Department of Physiology and Biophysics, Chicago, IL, USA; Magdi Yacoub Institute, Cardiac Biophysics Division, Harefield, UK; Imperial College, Heart and Lung Institute, London, UK; Freiburg University, Institute for Experimental Cardiovascular Medicine, Germany
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
23
|
Zhang H, Dvornikov AV, Huttner IG, Ma X, Santiago CF, Fatkin D, Xu X. A Langendorff-like system to quantify cardiac pump function in adult zebrafish. Dis Model Mech 2018; 11:dmm.034819. [PMID: 30012855 PMCID: PMC6177000 DOI: 10.1242/dmm.034819] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022] Open
Abstract
Zebrafish are increasingly used as a vertebrate model to study human cardiovascular disorders. Although heart structure and function are readily visualized in zebrafish embryos because of their optical transparency, the lack of effective tools for evaluating the hearts of older, nontransparent fish has been a major limiting factor. The recent development of high-frequency echocardiography has been an important advance for in vivo cardiac assessment, but it necessitates anesthesia and has limited ability to study acute interventions. We report the development of an alternative experimental ex vivo technique for quantifying heart size and function that resembles the Langendorff heart preparations that have been widely used in mammalian models. Dissected adult zebrafish hearts were perfused with a calcium-containing buffer, and a beat frequency was maintained with electrical stimulation. The impact of pacing frequency, flow rate and perfusate calcium concentration on ventricular performance (including end-diastolic and end-systolic volumes, ejection fraction, radial strain, and maximal velocities of shortening and relaxation) were evaluated and optimal conditions defined. We determined the effects of age on heart function in wild-type male and female zebrafish, and successfully detected hypercontractile and hypocontractile responses after adrenergic stimulation or doxorubicin treatment, respectively. Good correlations were found between indices of cardiac contractility obtained with high-frequency echocardiography and with the ex vivo technique in a subset of fish studied with both methods. The ex vivo beating heart preparation is a valuable addition to the cardiac function tool kit that will expand the use of adult zebrafish for cardiovascular research.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA.,Cardiovascular Surgery Department, the Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Alexey V Dvornikov
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| | - Inken G Huttner
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA.,Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55092, USA
| | - Celine F Santiago
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Diane Fatkin
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.,Cardiology Department, St. Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
24
|
Ma X, Ding Y, Wang Y, Xu X. A Doxorubicin-induced Cardiomyopathy Model in Adult Zebrafish. J Vis Exp 2018. [PMID: 29939187 DOI: 10.3791/57567] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The genetically accessible adult zebrafish (Danio rerio) has been increasingly used as a vertebrate model for understanding human diseases such as cardiomyopathy. Because of its convenience and amenability to high throughput genetic manipulations, the generation of acquired cardiomyopathy models, such as the doxorubicin-induced cardiomyopathy (DIC) model in adult zebrafish, is opening the doors to new research avenues, including discovering cardiomyopathy modifiers via forward genetic screening. Different from the embryonic zebrafish DIC model, both initial acute and later chronic phases of cardiomyopathy can be determined in the adult zebrafish DIC model, enabling the study of stage-dependent signaling mechanisms and therapeutic strategies. However, variable results can be obtained with the current model, even in the hands of experienced investigators. To facilitate future implementation of the DIC model, we present a detailed protocol on how to generate this DIC model in adult zebrafish and describe two alternative ways of intraperitoneal (IP) injection. We further discuss options on how to reduce variations to obtain reliable results and provide suggestions on how to appropriately interpret the results.
Collapse
Affiliation(s)
- Xiao Ma
- Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences; Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic
| | - Yong Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic; Institute of Life Science, Beijing University of Chinese Medicine
| | - Xiaolei Xu
- Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences; Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic;
| |
Collapse
|
25
|
Genome-wide association study of cardiotoxicity in the NCCTG N9831 (Alliance) adjuvant trastuzumab trial. Pharmacogenet Genomics 2018; 27:378-385. [PMID: 28763429 DOI: 10.1097/fpc.0000000000000302] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The major clinical side effect of the ERBB2-targeted breast cancer therapy, trastuzumab, is a decline in the left ventricular ejection fraction (LVEF). Improved markers are needed to better identify patients susceptible to cardiotoxicity. METHODS The NCCTG N9831 trial compared adjuvant doxorubicin and cyclophosphamide followed by either weekly paclitaxel (arm A); paclitaxel then trastuzumab (arm B); or concurrent paclitaxel and trastuzumab (arm C) in patients with HER2-positive breast cancer. A genome-wide association study was performed on all patients with available DNA (N=1446). We used linear regression to identify single nucleotide polymorphisms (SNPs) associated with decline in LVEF, adjusting for age, baseline LVEF, antihypertensive medications, and the first two principle components. RESULTS In total, 618 863 SNPs passed quality control and DNA from 1191 patients passed genotyping quality control and were identified as Whites of non-Hispanic origin. SNPs at six loci were associated with a decline in LVEF (P=7.73×10 to 8.93×10), LDB2, BRINP1, chr6 intergenic, RAB22A, TRPC6, and LINC01060, in patients who received chemotherapy plus trastuzumab (arms BC, N=800). None of these loci were significant in patients who received chemotherapy only (arm A, N=391) and did not increase in significance in the combined analysis of all patients. We did not observe association, P<0.05, with SNPs previously associated with trastuzumab-induced cardiotoxicity at ERBB2, I655V, and P1170A. We replicated association, P<0.05, with SNPs previously associated with anthracycline-induced cardiotoxicity at CBR3 and ABCB1. CONCLUSION Our study identified six putative novel cardiotoxicity loci in patients treated with combination chemotherapy and trastuzumab that require further investigation and confirmed known associations of anthracycline-induced cardiotoxicity.
Collapse
|
26
|
Meng E, Shevde LA, Samant RS. Emerging roles and underlying molecular mechanisms of DNAJB6 in cancer. Oncotarget 2018; 7:53984-53996. [PMID: 27276715 PMCID: PMC5288237 DOI: 10.18632/oncotarget.9803] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/26/2016] [Indexed: 12/29/2022] Open
Abstract
DNAJB6 also known as mammalian relative of DnaJ (MRJ) encodes a highly conserved member of the DnaJ/Hsp40 family of co-chaperone proteins that function with Hsp70 chaperones. DNAJB6 is widely expressed in all tissues, with higher expression levels detected in the brain. DNAJB6 is involved in diverse cellular functions ranging from murine placental development, reducing the formation and toxicity of mis-folded protein aggregates, to self-renewal of neural stem cells. Involvement of DNAJB6 is implicated in multiple pathologies such as Huntington's disease, Parkinson's diseases, limb-girdle muscular dystrophy, cardiomyocyte hypertrophy and cancer. This review summarizes the important involvement of the spliced isoforms of DNAJB6 in various pathologies with a specific focus on the emerging roles of human DNAJB6 in cancer and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Erhong Meng
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Beijing DOING Biomedical Technology Co. Ltd., Beijing,China
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
27
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
28
|
Tsai PC, Tsai YS, Soong BW, Huang YH, Wu HT, Chen YH, Lin KP, Liao YC, Lee YC. A novelDNAJB6mutation causes dominantly inherited distal-onset myopathy and compromises DNAJB6 function. Clin Genet 2017; 92:150-157. [DOI: 10.1111/cge.13001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022]
Affiliation(s)
- P.-C. Tsai
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| | - Y.-S. Tsai
- Center for Systems and Synthetic Biology; National Yang-Ming University; Taipei Taiwan
| | - B.-W. Soong
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| | - Y.-H. Huang
- Center for Systems and Synthetic Biology; National Yang-Ming University; Taipei Taiwan
- Institute of Biomedical Informatics; National Yang-Ming University; Taipei Taiwan
| | - H.-T. Wu
- Department of Radiology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Radiology; National Yang-Ming University School of Medicine; Taipei Taiwan
| | - Y.-H. Chen
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
| | - K.-P. Lin
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
| | - Y.-C. Liao
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
| | - Y.-C. Lee
- Department of Neurology; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Neurology; National Yang-Ming University School of Medicine; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
29
|
El-Rass S, Eisa-Beygi S, Khong E, Brand-Arzamendi K, Mauro A, Zhang H, Clark KJ, Ekker SC, Wen XY. Disruption of pdgfra alters endocardial and myocardial fusion during zebrafish cardiac assembly. Biol Open 2017; 6:348-357. [PMID: 28167492 PMCID: PMC5374395 DOI: 10.1242/bio.021212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiac development in vertebrates is a finely tuned process regulated by a set
of conserved signaling pathways. Perturbations of these processes are often
associated with congenital cardiac malformations. Platelet-derived growth factor
receptor α (PDGFRα) is a highly conserved tyrosine kinase
receptor, which is essential for development and organogenesis. Disruption of
Pdgfrα function in murine models is embryonic lethal
due to severe cardiovascular defects, suggesting a role in cardiac development,
thus necessitating the use of alternative models to explore its precise
function. In this study, we generated a zebrafish pdgfra mutant
line by gene trapping, in which the Pdgfra protein is truncated and fused with
mRFP (Pdgfra-mRFP). Our results demonstrate that pdgfra mutants
have defects in cardiac morphology as a result of abnormal fusion of myocardial
precursors. Expression analysis of the developing heart at later stages
suggested that Pdgfra-mRFP is expressed in the endocardium. Further examination
of the endocardium in pdgfra mutants revealed defective
endocardial migration to the midline, where cardiac fusion eventually occurs.
Together, our data suggests that pdgfra is required for proper
medial migration of both endocardial and myocardial precursors, an essential
step required for cardiac assembly and development. Summary: The molecular mechanisms regulating cardiac fusion are not
well understood. Here, we show that platelet-derived growth factor receptor
alpha is essential for normal endocardial and myocardial fusion during zebrafish
development.
Collapse
Affiliation(s)
- Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Collaborative Program in Cardiovascular Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2
| | - Shahram Eisa-Beygi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center. Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Edbert Khong
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8
| | - Koroboshka Brand-Arzamendi
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8
| | - Antonio Mauro
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Collaborative Program in Cardiovascular Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2
| | - Haibo Zhang
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Collaborative Program in Cardiovascular Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2.,Department of Medicine & Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada M5B 1T8 .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Collaborative Program in Cardiovascular Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2.,Department of Medicine & Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
30
|
Ding Y, Long PA, Bos JM, Shih YH, Ma X, Sundsbak RS, Chen J, Jiang Y, Zhao L, Hu X, Wang J, Shi Y, Ackerman MJ, Lin X, Ekker SC, Redfield MM, Olson TM, Xu X. A modifier screen identifies DNAJB6 as a cardiomyopathy susceptibility gene. JCI Insight 2016; 1. [PMID: 27642634 DOI: 10.1172/jci.insight.88797] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mutagenesis screening is a powerful forward genetic approach that has been successfully applied in lower-model organisms to discover genetic factors for biological processes. This phenotype-based approach has yet to be established in vertebrates for probing major human diseases, largely because of the complexity of colony management. Herein, we report a rapid strategy for identifying genetic modifiers of cardiomyopathy (CM). Based on the application of doxorubicin stress to zebrafish insertional cardiac (ZIC) mutants, we identified 4 candidate CM-modifying genes, of which 3 have been linked previously to CM. The long isoform of DnaJ (Hsp40) homolog, subfamily B, member 6b (dnajb6b(L)) was identified as a CM susceptibility gene, supported by identification of rare variants in its human ortholog DNAJB6 from CM patients. Mechanistic studies indicated that the deleterious, loss-of-function modifying effects of dnajb6b(L) can be ameliorated by inhibition of ER stress. In contrast, overexpression of dnajb6(L) exerts cardioprotective effects on both fish and mouse CM models. Together, our findings establish a mutagenesis screening strategy that is scalable for systematic identification of genetic modifiers of CM, feasible to suggest therapeutic targets, and expandable to other major human diseases.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Pamela A Long
- Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - J Martijn Bos
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yu-Huan Shih
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Rhianna S Sundsbak
- Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwen Jiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Jianan Wang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Yongyong Shi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Michael J Ackerman
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Timothy M Olson
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA; Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
31
|
St Hilaire C, Liberman M, Miller JD. Bidirectional Translation in Cardiovascular Calcification. Arterioscler Thromb Vasc Biol 2016; 36:e19-24. [PMID: 26912744 DOI: 10.1161/atvbaha.115.307056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Cynthia St Hilaire
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | - Marcel Liberman
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | - Jordan D Miller
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | | |
Collapse
|
32
|
Bellipanni G, Cappello F, Scalia F, Conway de Macario E, Macario AJ, Giordano A. Zebrafish as a Model for the Study of Chaperonopathies. J Cell Physiol 2016; 231:2107-14. [DOI: 10.1002/jcp.25319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Gianfranco Bellipanni
- Sbarro Institute for Cancer Research and Molecular Medicine; Philadelphia Pennsylvania
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
| | - Francesco Cappello
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
- Department of Experimental Biomedicine and Clinical Neuroscience; University of Palermo; Palermo Italy
| | - Federica Scalia
- Department of Experimental Biomedicine and Clinical Neuroscience; University of Palermo; Palermo Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology; School of Medicine, University of Maryland at Baltimore and IMET; Baltimore Maryland
| | - Alberto J.L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
- Department of Microbiology and Immunology; School of Medicine, University of Maryland at Baltimore and IMET; Baltimore Maryland
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine; Philadelphia Pennsylvania
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
| |
Collapse
|
33
|
Westcot SE, Hatzold J, Urban MD, Richetti SK, Skuster KJ, Harm RM, Lopez Cervera R, Umemoto N, McNulty MS, Clark KJ, Hammerschmidt M, Ekker SC. Protein-Trap Insertional Mutagenesis Uncovers New Genes Involved in Zebrafish Skin Development, Including a Neuregulin 2a-Based ErbB Signaling Pathway Required during Median Fin Fold Morphogenesis. PLoS One 2015; 10:e0130688. [PMID: 26110643 PMCID: PMC4482254 DOI: 10.1371/journal.pone.0130688] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/24/2015] [Indexed: 01/13/2023] Open
Abstract
Skin disorders are widespread, but available treatments are limited. A more comprehensive understanding of skin development mechanisms will drive identification of new treatment targets and modalities. Here we report the Zebrafish Integument Project (ZIP), an expression-driven platform for identifying new skin genes and phenotypes in the vertebrate model Danio rerio (zebrafish). In vivo selection for skin-specific expression of gene-break transposon (GBT) mutant lines identified eleven new, revertible GBT alleles of genes involved in skin development. Eight genes—fras1, grip1, hmcn1, msxc, col4a4, ahnak, capn12, and nrg2a—had been described in an integumentary context to varying degrees, while arhgef25b, fkbp10b, and megf6a emerged as novel skin genes. Embryos homozygous for a GBT insertion within neuregulin 2a (nrg2a) revealed a novel requirement for a Neuregulin 2a (Nrg2a) – ErbB2/3 – AKT signaling pathway governing the apicobasal organization of a subset of epidermal cells during median fin fold (MFF) morphogenesis. In nrg2a mutant larvae, the basal keratinocytes within the apical MFF, known as ridge cells, displayed reduced pAKT levels as well as reduced apical domains and exaggerated basolateral domains. Those defects compromised proper ridge cell elongation into a flattened epithelial morphology, resulting in thickened MFF edges. Pharmacological inhibition verified that Nrg2a signals through the ErbB receptor tyrosine kinase network. Moreover, knockdown of the epithelial polarity regulator and tumor suppressor lgl2 ameliorated the nrg2a mutant phenotype. Identifying Lgl2 as an antagonist of Nrg2a – ErbB signaling revealed a significantly earlier role for Lgl2 during epidermal morphogenesis than has been described to date. Furthermore, our findings demonstrated that successive, coordinated ridge cell shape changes drive apical MFF development, making MFF ridge cells a valuable model for investigating how the coordinated regulation of cell polarity and cell shape changes serves as a crucial mechanism of epithelial morphogenesis.
Collapse
Affiliation(s)
- Stephanie E. Westcot
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Julia Hatzold
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mark D. Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stefânia K. Richetti
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
| | - Kimberly J. Skuster
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Rhianna M. Harm
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Roberto Lopez Cervera
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Noriko Umemoto
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Melissa S. McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karl J. Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthias Hammerschmidt
- Institute for Developmental Biology, University of Cologne, Biocenter, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Stephen C. Ekker
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
34
|
Yang J, Shih YH, Xu X. Understanding cardiac sarcomere assembly with zebrafish genetics. Anat Rec (Hoboken) 2015; 297:1681-93. [PMID: 25125181 DOI: 10.1002/ar.22975] [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: 01/08/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 01/06/2023]
Abstract
Mutations in sarcomere genes have been found in many inheritable human diseases, including hypertrophic cardiomyopathy. Elucidating the molecular mechanisms of sarcomere assembly shall facilitate understanding of the pathogenesis of sarcomere-based cardiac disease. Recently, biochemical and genomic studies have identified many new genes encoding proteins that localize to the sarcomere. However, their precise functions in sarcomere assembly and sarcomere-based cardiac disease are unknown. Here, we review zebrafish as an emerging vertebrate model for these studies. We summarize the techniques offered by this animal model to manipulate genes of interest, annotate gene expression, and describe the resulting phenotypes. We survey the sarcomere genes that have been investigated in zebrafish and discuss the potential of applying this in vivo model for larger-scale genetic studies.
Collapse
Affiliation(s)
- Jingchun Yang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota; Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | | |
Collapse
|
35
|
Shih YH, Zhang Y, Ding Y, Ross CA, Li H, Olson TM, Xu X. Cardiac transcriptome and dilated cardiomyopathy genes in zebrafish. ACTA ACUST UNITED AC 2015; 8:261-9. [PMID: 25583992 DOI: 10.1161/circgenetics.114.000702] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 12/16/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genetic studies of cardiomyopathy and heart failure have limited throughput in mammalian models. Adult zebrafish have been recently pursued as a vertebrate model with higher throughput, but genetic conservation must be tested. METHODS AND RESULTS We conducted transcriptome analysis of zebrafish heart and searched for fish homologues of 51 known human dilated cardiomyopathy-associated genes. We also identified genes with high cardiac expression and genes with differential expression between embryonic and adult stages. Among tested genes, 30 had a single zebrafish orthologue, 14 had 2 homologues, and 5 had ≥3 homologues. By analyzing the expression data on the basis of cardiac abundance and enrichment hypotheses, we identified a single zebrafish gene for 14 of 19 multiple-homologue genes and 2 zebrafish homologues of high priority for ACTC1. Of note, our data suggested vmhc and vmhcl as functional zebrafish orthologues for human genes MYH6 and MYH7, respectively, which are established molecular markers for cardiac remodeling. CONCLUSIONS Most known genes for human dilated cardiomyopathy have a corresponding zebrafish orthologue, which supports the use of zebrafish as a conserved vertebrate model. Definition of the cardiac transcriptome and fetal gene program will facilitate systems biology studies of dilated cardiomyopathy in zebrafish.
Collapse
Affiliation(s)
- Yu-Huan Shih
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Yuji Zhang
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Yonghe Ding
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Christian A Ross
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Hu Li
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Timothy M Olson
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.)
| | - Xiaolei Xu
- From the Department of Biochemistry and Molecular Biology (Y.-H.S., Y.D., X.X.), Information Technology (C.A.R.), Department of Molecular Pharmacology and Experimental Therapeutics (H.L.), Department of Pediatric and Adolescent Medicine (T.M.O.), and Division of Cardiovascular Diseases (T.M.O., X.X.), Mayo Clinic, Rochester, MN; Division of Biostatistics and Bioinformatics, University of Maryland Greenebaum Cancer Center, Baltimore (Y.Z.); and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore (Y.Z.).
| |
Collapse
|
36
|
Guo DC, Gong L, Regalado E, Santos-Cortez R, Zhao R, Cai B, Veeraraghavan S, Prakash S, Johnson R, Muilenburg A, Willing M, Jondeau G, Boileau C, Pannu H, Moran R, Debacker J, Bamshad M, Shendure J, Nickerson D, Leal S, Raman C, Swindell E, Milewicz D, Swindell EC, Milewicz DM. MAT2A mutations predispose individuals to thoracic aortic aneurysms. Am J Hum Genet 2015; 96:170-7. [PMID: 25557781 DOI: 10.1016/j.ajhg.2014.11.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/30/2014] [Indexed: 02/06/2023] Open
Abstract
Up to 20% of individuals who have thoracic aortic aneurysms or acute aortic dissections but who do not have syndromic features have a family history of thoracic aortic disease. Significant genetic heterogeneity is established for this familial condition. Whole-genome linkage analysis and exome sequencing of distant relatives from a large family with autosomal-dominant inheritance of thoracic aortic aneurysms variably associated with the bicuspid aortic valve was used for identification of additional genes predisposing individuals to this condition. A rare variant, c.1031A>C (p.Glu344Ala), was identified in MAT2A, which encodes methionine adenosyltransferase II alpha (MAT IIα). This variant segregated with disease in the family, and Sanger sequencing of DNA from affected probands from unrelated families with thoracic aortic disease identified another MAT2A rare variant, c.1067G>A (p.Arg356His). Evidence that these variants predispose individuals to thoracic aortic aneurysms and dissections includes the following: there is a paucity of rare variants in MAT2A in the population; amino acids Glu344 and Arg356 are conserved from humans to zebrafish; and substitutions of these amino acids in MAT Iα are found in individuals with hypermethioninemia. Structural analysis suggested that p.Glu344Ala and p.Arg356His disrupt MAT IIα enzyme function. Knockdown of mat2aa in zebrafish via morpholino oligomers disrupted cardiovascular development. Co-transfected wild-type human MAT2A mRNA rescued defects of zebrafish cardiovascular development at significantly higher levels than mRNA edited to express either the Glu344 or Arg356 mutants, providing further evidence that the p.Glu344Ala and p.Arg356His substitutions impair MAT IIα function. The data presented here support the conclusion that rare genetic variants in MAT2A predispose individuals to thoracic aortic disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric C Swindell
- Department of Pediatrics, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Dianna M Milewicz
- Department of Internal Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
37
|
Opitz R, Antonica F, Costagliola S. New model systems to illuminate thyroid organogenesis. Part I: an update on the zebrafish toolbox. Eur Thyroid J 2013; 2:229-42. [PMID: 24783054 PMCID: PMC3923603 DOI: 10.1159/000357079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/07/2013] [Indexed: 12/16/2022] Open
Abstract
Thyroid dysgenesis (TD) resulting from defects during embryonic thyroid development represents a major cause of congenital hypothyroidism. The pathogenetic mechanisms of TD in human newborns, however, are still poorly understood and disease-causing genetic variants have been identified in only a small percentage of TD cases. This limited understanding of the pathogenesis of TD is partly due to a lack of knowledge on how intrinsic factors and extrinsic signalling cues orchestrate the differentiation of thyroid follicular cells and the morphogenesis of thyroid tissue. Recently, embryonic stem cells and zebrafish embryos emerged as novel model systems that allow for innovative experimental approaches in order to decipher cellular and molecular mechanisms of thyroid development and to unravel pathogenic mechanisms of TD. Zebrafish embryos offer several salient properties for studies on thyroid organogenesis including rapid and external development, optical transparency, ease of breeding, relative short generation time and amenability for genome editing. In this review, we will highlight recent advances in the zebrafish toolkit to visualize cellular dynamics of organ development and discuss specific prospects of the zebrafish model for studies on vertebrate thyroid development and human congenital thyroid diseases.
Collapse
Affiliation(s)
- Robert Opitz
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Francesco Antonica
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
38
|
Varshney GK, Burgess SM. Mutagenesis and phenotyping resources in zebrafish for studying development and human disease. Brief Funct Genomics 2013; 13:82-94. [PMID: 24162064 DOI: 10.1093/bfgp/elt042] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The zebrafish (Danio rerio) is an important model organism for studying development and human disease. The zebrafish has an excellent reference genome and the functions of hundreds of genes have been tested using both forward and reverse genetic approaches. Recent years have seen an increasing number of large-scale mutagenesis projects and the number of mutants or gene knockouts in zebrafish has increased rapidly, including for the first time conditional knockout technologies. In addition, targeted mutagenesis techniques such as zinc finger nucleases, transcription activator-like effector nucleases and clustered regularly interspaced short sequences (CRISPR) or CRISPR-associated (Cas), have all been shown to effectively target zebrafish genes as well as the first reported germline homologous recombination, further expanding the utility and power of zebrafish genetics. Given this explosion of mutagenesis resources, it is now possible to perform systematic, high-throughput phenotype analysis of all zebrafish gene knockouts.
Collapse
Affiliation(s)
- Gaurav Kumar Varshney
- Developmental Genomics Section, Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
39
|
Song G, Cui Z. Novel strategies for gene trapping and insertional mutagenesis mediated by Sleeping Beauty transposon. Mob Genet Elements 2013; 3:e26499. [PMID: 24251071 DOI: 10.4161/mge.26499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 09/10/2013] [Accepted: 09/15/2013] [Indexed: 12/29/2022] Open
Abstract
Gene and poly(A) trappings are high-throughput approaches to capture and interrupt the expression of endogenous genes within a target genome. Although a number of trapping vectors have been developed for investigation of gene functions in cells and vertebrate models, there is still room for the improvement of their efficiency and sensitivity. Recently, two novel trapping vectors mediated by Sleeping Beauty (SB) transposon have been generated by the combination of three functional cassettes that are required for finding endogenous genes, disrupting the expression of trapped genes, and inducing the excision of integrated traps from their original insertion sites and then inserting into another gene. In addition, several other strategies are utilized to improve the activities of two trapping vectors. First, activities of all components were examined in vitro before the generation of two vectors. Second, the inducible promoter from the tilapia Hsp70 gene was used to drive the expression of SB gene, which can mediate the excision of integrated transposons upon induction at 37 °C. Third, the Cre/LoxP system was introduced to delete the SB expression cassette for stabilization of gene interruption and bio-safety. Fourth, three stop codons in different reading frames were introduced downstream of a strong splice acceptor (SA) in the gene trapping vector to effectively terminate the translation of trapped endogenous genes. Fifth, the strong splicing donor (SD) and AU-rich RNA-destabilizing element exhibited no obvious insertion bias and markedly reduced SD read-through events, and the combination of an enhanced SA, a poly(A) signal and a transcript terminator in the poly(A) trapping vector efficiently disrupted the transcription of trapped genes. Thus, these two trapping vectors are alternative and effective tools for large-scale identification and disruption of endogenous genes in vertebrate cells and animals.
Collapse
Affiliation(s)
- Guili Song
- Institute of Hydrobiology; Chinese Academy of Sciences; Wuhan, P.R. China
| | | |
Collapse
|