1
|
Liu N, Nakano H, Nakano A. Mutual Regulation of Cardiovascular and Hematopoietic Development. Curr Cardiol Rep 2025; 27:86. [PMID: 40261519 PMCID: PMC12014711 DOI: 10.1007/s11886-025-02236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW The cardiovascular and hematopoietic systems share molecular mechanisms and regulatory interactions across species. Endocardial hematopoiesis, a debated topic in mice, is actually an evolutionarily conserved process from Drosophila. This review explores the origins and significance of endocardial hematopoiesis, highlighting its role in cardiac development and macrophage formation. RECENT FINDINGS Despite extensive lineage-tracing and transcriptome studies, it remained unclear until single-cell RNA sequencing (scRNA-seq) identified that endocardial cells possess an intrinsic hematopoietic program independent of known hematopoietic organs. These endocardial-derived macrophages contribute uniquely to cardiac morphogenesis, supporting valve maturation and tissue remodeling. Endocardial hematopoiesis is an evolutionarily conserved phenomenon that is essential for developmental process. The heterogeneity of tissue-resident macrophages and their specialized functions in cardiac development have been further unraveled by single-cell analysis. This review provides an evolutionary perspective on endocardial hematopoiesis and highlights its critical contributions of hematopoietic cells to heart formation and homeostasis.
Collapse
Affiliation(s)
- Norika Liu
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjyo, Chuou-ku, Kumamoto-Shi, Kumamoto, 860-0811, Japan.
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, USA.
| | - Haruko Nakano
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, USA
| | - Atsushi Nakano
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, USA
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- David Geffen Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA
| |
Collapse
|
2
|
Farmer AJ, Katariya R, Islam S, Rayhan MSA, Inlow MH, Ahmad SM, Schwab KR. trithorax is an essential regulator of cardiac Hox gene expression and anterior-posterior patterning of the Drosophila embryonic heart tube. Biol Open 2025; 14:bio061919. [PMID: 40172069 PMCID: PMC11993250 DOI: 10.1242/bio.061919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 04/04/2025] Open
Abstract
The precise regulation of transcription required for embryonic development is partially controlled by the actions of the Trithorax group (TrxG) and Polycomb group (PcG) proteins. The genes trithorax (trx), trithorax-related (trr), and SET domain containing 1 (Set1) encode COMPASS-like histone methyltransferases, a subgroup of TrxG proteins that impart H3K4 methylation modifications onto chromatin in order to activate and maintain transcription. In this study, we identify the role of these genes in the development of the embryonic heart of the fruit fly Drosophila melanogaster. trx, trr, and Set1 independently ensure proper cardiac cell divisions. Additionally, trx regulation of collinear Hox expression is necessary for the anterior-posterior cardiac patterning of the linear heart tube. trx inactivation in Drosophila results in a remarkable homeotic transformation of the posterior heart-proper segment into an aorta-like fate due to the loss of posterior abdominal A expression. Furthermore, cardiac expression of Antennapedia, Ultrabithorax, and Abdominal B is also deregulated in trx mutants. Together, these data suggest that the COMPASS-like histone methyltransferases are essential developmental regulators of cardiogenesis, being necessary for both cardiac cell divisions and heart patterning.
Collapse
Affiliation(s)
- Adam J. Farmer
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Rajnandani Katariya
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Sumaiya Islam
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Md. Sayeed Abu Rayhan
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Mark H. Inlow
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Department of Mathematics and Computer Science, Indiana State University, Terre Haute, IN 47809, USA
| | - Shaad M. Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kristopher R. Schwab
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| |
Collapse
|
3
|
Hasan MR, Kump AJ, Stepaniak EC, Panta M, Shashidhar K, Katariya R, Sabbir MK, Schwab KR, Inlow MH, Chen Y, Ahmad SM. Genome-Wide Expression Profiling and Phenotypic Analysis of Downstream Targets Identify the Fox Transcription Factor Jumeau as a Master Regulator of Cardiac Progenitor Cell Division. Int J Mol Sci 2024; 25:12933. [PMID: 39684645 DOI: 10.3390/ijms252312933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Forkhead box (Fox) transcription factors (TFs) mediate multiple conserved cardiogenic processes in both mammals and Drosophila. Our prior work identified the roles of two Drosophila Fox genes, jumeau (jumu) and Checkpoint suppressor 1-like (CHES-1-like), in cardiac progenitor cell specification and division, and in the proper positioning of cardiac cell subtypes. Fox TF binding sites are also significantly enriched in the enhancers of genes expressed in the heart, suggesting that these genes may play a core regulatory role in one or more of these cardiogenic processes. We identified downstream targets of Jumu by comparing transcriptional expression profiles of flow cytometry-sorted mesodermal cells from wild-type embryos and embryos completely lacking the jumu gene and found that genes with functional annotation and ontological features suggesting roles in cell division were overrepresented among Jumu targets. Phenotypic analysis of a subset of these targets identified 21 jumu-regulated genes that mediate cardiac progenitor cell division, one of which, Retinal Homeobox (Rx), was characterized in more detail. Finally, the observation that many of these 21 genes and/or their orthologs exhibit genetic or physical interactions among themselves indicates that Jumu is a master regulator acting as a hub of a cardiac progenitor cell division-mediating network.
Collapse
Affiliation(s)
- M Rezaul Hasan
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Andrew J Kump
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Evelyn C Stepaniak
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manoj Panta
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kuncha Shashidhar
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Rajnandani Katariya
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mofazzal K Sabbir
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kristopher R Schwab
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mark H Inlow
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Department of Mathematical Sciences, Indiana State University, Terre Haute, IN 47809, USA
| | - Ye Chen
- Department of Mathematics and Statistics, Northern Arizona University, Flagstaff, AZ 86001, USA
| | - Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| |
Collapse
|
4
|
Pallarès-Albanell J, Ortega-Flores L, Senar-Serra T, Ruiz A, Abril JF, Rossello M, Almudi I. Gene regulatory dynamics during the development of a paleopteran insect, the mayfly Cloeon dipterum. Development 2024; 151:dev203017. [PMID: 39324209 PMCID: PMC11491810 DOI: 10.1242/dev.203017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
The evolution of insects has been marked by the appearance of key body plan innovations that promoted the outstanding ability of this lineage to adapt to new habitats, boosting the most successful radiation in animals. To understand the evolution of these new structures, it is essential to investigate which genes and gene regulatory networks participate during the embryonic development of insects. Great efforts have been made to fully understand gene expression and gene regulation during the development of holometabolous insects, in particular Drosophila melanogaster. Conversely, functional genomics resources and databases in other insect lineages are scarce. To provide a new platform to study gene regulation in insects, we generated ATAC-seq for the first time during the development of the mayfly Cloeon dipterum, which belongs to Paleoptera, the sister group to all other winged insects. With these comprehensive datasets along six developmental stages, we characterized pronounced changes in accessible chromatin between early and late embryogenesis. The application of ATAC-seq in mayflies provides a fundamental resource to understand the evolution of gene regulation in insects.
Collapse
Affiliation(s)
- Joan Pallarès-Albanell
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Laia Ortega-Flores
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Tòt Senar-Serra
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Antoni Ruiz
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Josep F. Abril
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institute of Biomedicine of Universitat de Barcelona (IBUB), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Maria Rossello
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Isabel Almudi
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain
| |
Collapse
|
5
|
Sloutskin A, Itzhak D, Vogler G, Pozeilov H, Ideses D, Alter H, Adato O, Shachar H, Doniger T, Shohat-Ophir G, Frasch M, Bodmer R, Duttke SH, Juven-Gershon T. From promoter motif to cardiac function: a single DPE motif affects transcription regulation and organ function in vivo. Development 2024; 151:dev202355. [PMID: 38958007 DOI: 10.1242/dev.202355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
Transcription initiates at the core promoter, which contains distinct core promoter elements. Here, we highlight the complexity of transcriptional regulation by outlining the effect of core promoter-dependent regulation on embryonic development and the proper function of an organism. We demonstrate in vivo the importance of the downstream core promoter element (DPE) in complex heart formation in Drosophila. Pioneering a novel approach using both CRISPR and nascent transcriptomics, we show the effects of mutating a single core promoter element within the natural context. Specifically, we targeted the downstream core promoter element (DPE) of the endogenous tin gene, encoding the Tinman transcription factor, a homologue of human NKX2-5 associated with congenital heart diseases. The 7 bp substitution mutation results in massive perturbation of the Tinman regulatory network that orchestrates dorsal musculature, which is manifested as physiological and anatomical changes in the cardiac system, impaired specific activity features, and significantly compromised viability of adult flies. Thus, a single motif can have a critical impact on embryogenesis and, in the case of DPE, functional heart formation.
Collapse
Affiliation(s)
- Anna Sloutskin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dekel Itzhak
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hadar Pozeilov
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Diana Ideses
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Hadar Alter
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Orit Adato
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Hadar Shachar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tirza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Galit Shohat-Ophir
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Manfred Frasch
- Division of Developmental Biology, Department of Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sascha H Duttke
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Tamar Juven-Gershon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
6
|
Bileckyj C, Blotz B, Cripps RM. Drosophila as a Model to Understand Second Heart Field Development. J Cardiovasc Dev Dis 2023; 10:494. [PMID: 38132661 PMCID: PMC10744189 DOI: 10.3390/jcdd10120494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The genetic model system Drosophila has contributed fundamentally to our understanding of mammalian heart specification, development, and congenital heart disease. The relatively simple Drosophila heart is a linear muscular tube that is specified and develops in the embryo and persists throughout the life of the animal. It functions at all stages to circulate hemolymph within the open circulatory system of the body. During Drosophila metamorphosis, the cardiac tube is remodeled, and a new layer of muscle fibers spreads over the ventral surface of the heart to form the ventral longitudinal muscles. The formation of these fibers depends critically upon genes known to be necessary for mammalian second heart field (SHF) formation. Here, we review the prior contributions of the Drosophila system to the understanding of heart development and disease, discuss the importance of the SHF to mammalian heart development and disease, and then discuss how the ventral longitudinal adult cardiac muscles can serve as a novel model for understanding SHF development and disease.
Collapse
Affiliation(s)
| | | | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
7
|
Lovato TL, Blotz B, Bileckyj C, Johnston CA, Cripps RM. Modeling a variant of unknown significance in the Drosophila ortholog of the human cardiogenic gene NKX2.5. Dis Model Mech 2023; 16:dmm050059. [PMID: 37691628 PMCID: PMC10548113 DOI: 10.1242/dmm.050059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
Sequencing of human genome samples has unearthed genetic variants for which functional testing is necessary to validate their clinical significance. We used the Drosophila system to analyze a variant of unknown significance in the human congenital heart disease gene NKX2.5 (also known as NKX2-5). We generated an R321N allele of the NKX2.5 ortholog tinman (tin) to model a human K158N variant and tested its function in vitro and in vivo. The R321N Tin isoform bound poorly to DNA in vitro and was deficient in activating a Tin-dependent enhancer in tissue culture. Mutant Tin also showed a significantly reduced interaction with a Drosophila T-box cardiac factor named Dorsocross1. We generated a tinR321N allele using CRISPR/Cas9, for which homozygotes were viable and had normal heart specification, but showed defects in the differentiation of the adult heart that were exacerbated by further loss of tin function. We propose that the human K158N variant is pathogenic through causing a deficiency in DNA binding and a reduced ability to interact with a cardiac co-factor, and that cardiac defects might arise later in development or adult life.
Collapse
Affiliation(s)
- TyAnna L. Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Brenna Blotz
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Cayleen Bileckyj
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | | | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
8
|
Kervadec A, Kezos J, Ni H, Yu M, Marchant J, Spiering S, Kannan S, Kwon C, Andersen P, Bodmer R, Grandi E, Ocorr K, Colas AR. Multiplatform modeling of atrial fibrillation identifies phospholamban as a central regulator of cardiac rhythm. Dis Model Mech 2023; 16:dmm049962. [PMID: 37293707 PMCID: PMC10387351 DOI: 10.1242/dmm.049962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
Atrial fibrillation (AF) is a common and genetically inheritable form of cardiac arrhythmia; however, it is currently not known how these genetic predispositions contribute to the initiation and/or maintenance of AF-associated phenotypes. One major barrier to progress is the lack of experimental systems to investigate the effects of gene function on rhythm parameters in models with human atrial and whole-organ relevance. Here, we assembled a multi-model platform enabling high-throughput characterization of the effects of gene function on action potential duration and rhythm parameters using human induced pluripotent stem cell-derived atrial-like cardiomyocytes and a Drosophila heart model, and validation of the findings using computational models of human adult atrial myocytes and tissue. As proof of concept, we screened 20 AF-associated genes and identified phospholamban loss of function as a top conserved hit that shortens action potential duration and increases the incidence of arrhythmia phenotypes upon stress. Mechanistically, our study reveals that phospholamban regulates rhythm homeostasis by functionally interacting with L-type Ca2+ channels and NCX. In summary, our study illustrates how a multi-model system approach paves the way for the discovery and molecular delineation of gene regulatory networks controlling atrial rhythm with application to AF.
Collapse
Affiliation(s)
- Anaïs Kervadec
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Haibo Ni
- Department of Pharmacology, UC Davis, Davis, CA 95616, USA
| | - Michael Yu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Marchant
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sean Spiering
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Suraj Kannan
- Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Rolf Bodmer
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexandre R. Colas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
9
|
Lovato TL, Blotz B, Bileckyj C, Johnston CA, Cripps RM. Using Drosophila to model a variant of unknown significance in the human cardiogenic gene Nkx2.5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546937. [PMID: 37425758 PMCID: PMC10327092 DOI: 10.1101/2023.06.28.546937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Sequencing of human genome samples has unearthed genetic variants for which functional testing is necessary to validate their clinical significance. We used the Drosophila system to analyze a variant of unknown significance in the human congenital heart disease gene, Nkx2 . 5 . We generated an R321N allele of the Nkx2 . 5 ortholog tinman ( tin ) to model a human K158N variant and tested its function in vitro and in vivo. The R321N Tin isoform bound poorly to DNA in vitro and was deficient in activating a Tin-dependent enhancer in tissue culture. Mutant Tin also showed a significantly reduced interaction with a Drosophila Tbox cardiac factor named Dorsocross1. We generated a tin R321N allele using CRISPR/Cas9, for which homozygotes were viable and had normal heart specification, but showed defects in the differentiation of the adult heart that were exacerbated by further loss of tin function. We conclude that the human K158N mutation is likely pathogenic through causing both a deficiency in DNA binding and a reduced ability to interact with a cardiac cofactor, and that cardiac defects might arise later in development or adult life.
Collapse
|
10
|
Sloutskin A, Itzhak D, Vogler G, Ideses D, Alter H, Shachar H, Doniger T, Frasch M, Bodmer R, Duttke SH, Juven-Gershon T. A single DPE core promoter motif contributes to in vivo transcriptional regulation and affects cardiac function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.11.544490. [PMID: 37398300 PMCID: PMC10312617 DOI: 10.1101/2023.06.11.544490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Transcription is initiated at the core promoter, which confers specific functions depending on the unique combination of core promoter elements. The downstream core promoter element (DPE) is found in many genes related to heart and mesodermal development. However, the function of these core promoter elements has thus far been studied primarily in isolated, in vitro or reporter gene settings. tinman (tin) encodes a key transcription factor that regulates the formation of the dorsal musculature and heart. Pioneering a novel approach utilizing both CRISPR and nascent transcriptomics, we show that a substitution mutation of the functional tin DPE motif within the natural context of the core promoter results in a massive perturbation of Tinman's regulatory network orchestrating dorsal musculature and heart formation. Mutation of endogenous tin DPE reduced the expression of tin and distinct target genes, resulting in significantly reduced viability and an overall decrease in adult heart function. We demonstrate the feasibility and importance of characterizing DNA sequence elements in vivo in their natural context, and accentuate the critical impact a single DPE motif has during Drosophila embryogenesis and functional heart formation.
Collapse
Affiliation(s)
- Anna Sloutskin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Dekel Itzhak
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Diana Ideses
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Alter
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Shachar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tirza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Manfred Frasch
- Division of Developmental Biology, Department of Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sascha H Duttke
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Tamar Juven-Gershon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
11
|
Gao X, Yan B. The mechanism and diagnostic value of Tbx20 in cardiovascular diseases. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
12
|
McAndry C, Collins M, Tills O, Spicer JI, Truebano M. Regulation of gene expression during ontogeny of physiological function in the brackishwater amphipod Gammarus chevreuxi. Mar Genomics 2022; 63:100948. [PMID: 35427917 DOI: 10.1016/j.margen.2022.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
Embryonic development is a complex process involving the co-ordinated onset and integration of multiple morphological features and physiological functions. While the molecular basis of morphological development in embryos is relatively well known for traditional model species, the molecular underpinning of the development of physiological functions is not. Here, we used global gene expression profiling to investigate the transcriptional changes associated with the development of morphological and physiological function in the amphipod crustacean Gammarus chevreuxi. We compared the transcriptomes at three timepoints during the latter half of development, characterised by different stages of the development of heart form and function: 10 days post fertilisation (dpf, Early: no heart structure visible), 15 dpf (Middle: heart present but not fully functional), and 18 dpf (Late: regular heartbeat). Gene expression profiles differed markedly between developmental stages, likely representing a change in the activity of different processes throughout the latter period of G. chevreuxi embryonic development. Differentially expressed genes belonged to one of three distinct clusters based on their expression patterns across development. One of these clusters, which included key genes relating to cardiac contractile machinery and calcium handling, displayed a pattern of sequential up-regulation throughout the developmental period studied. Further analyses of these transcripts could reveal genes that may influence the onset of a regular heartbeat. We also identified morphological and physiological processes that may occur alongside heart development, such as development of digestive caeca and the cuticle. Elucidating the mechanisms underpinning morphological and physiological development of non-model organisms will support improved understanding of conserved mechanisms, addressing the current phylogenetic gap between relatively well known model species.
Collapse
Affiliation(s)
- C McAndry
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - M Collins
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - O Tills
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - J I Spicer
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - M Truebano
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK.
| |
Collapse
|
13
|
Rice C, De O, Alhadyian H, Hall S, Ward RE. Expanding the Junction: New Insights into Non-Occluding Roles for Septate Junction Proteins during Development. J Dev Biol 2021; 9:11. [PMID: 33801162 PMCID: PMC8006247 DOI: 10.3390/jdb9010011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
The septate junction (SJ) provides an occluding function for epithelial tissues in invertebrate organisms. This ability to seal the paracellular route between cells allows internal tissues to create unique compartments for organ function and endows the epidermis with a barrier function to restrict the passage of pathogens. Over the past twenty-five years, numerous investigators have identified more than 30 proteins that are required for the formation or maintenance of the SJs in Drosophila melanogaster, and have determined many of the steps involved in the biogenesis of the junction. Along the way, it has become clear that SJ proteins are also required for a number of developmental events that occur throughout the life of the organism. Many of these developmental events occur prior to the formation of the occluding junction, suggesting that SJ proteins possess non-occluding functions. In this review, we will describe the composition of SJs, taking note of which proteins are core components of the junction versus resident or accessory proteins, and the steps involved in the biogenesis of the junction. We will then elaborate on the functions that core SJ proteins likely play outside of their role in forming the occluding junction and describe studies that provide some cell biological perspectives that are beginning to provide mechanistic understanding of how these proteins function in developmental contexts.
Collapse
Affiliation(s)
- Clinton Rice
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA; (C.R.); (H.A.)
| | - Oindrila De
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Haifa Alhadyian
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA; (C.R.); (H.A.)
| | | | - Robert E. Ward
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA;
| |
Collapse
|
14
|
Theis JL, Vogler G, Missinato MA, Li X, Nielsen T, Zeng XXI, Martinez-Fernandez A, Walls SM, Kervadec A, Kezos JN, Birker K, Evans JM, O'Byrne MM, Fogarty ZC, Terzic A, Grossfeld P, Ocorr K, Nelson TJ, Olson TM, Colas AR, Bodmer R. Patient-specific genomics and cross-species functional analysis implicate LRP2 in hypoplastic left heart syndrome. eLife 2020; 9:e59554. [PMID: 33006316 PMCID: PMC7581429 DOI: 10.7554/elife.59554] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Congenital heart diseases (CHDs), including hypoplastic left heart syndrome (HLHS), are genetically complex and poorly understood. Here, a multidisciplinary platform was established to functionally evaluate novel CHD gene candidates, based on whole-genome and iPSC RNA sequencing of a HLHS family-trio. Filtering for rare variants and altered expression in proband iPSCs prioritized 10 candidates. siRNA/RNAi-mediated knockdown in healthy human iPSC-derived cardiomyocytes (hiPSC-CM) and in developing Drosophila and zebrafish hearts revealed that LDL receptor-related protein LRP2 is required for cardiomyocyte proliferation and differentiation. Consistent with hypoplastic heart defects, compared to patents the proband's iPSC-CMs exhibited reduced proliferation. Interestingly, rare, predicted-damaging LRP2 variants were enriched in a HLHS cohort; however, understanding their contribution to HLHS requires further investigation. Collectively, we have established a multi-species high-throughput platform to rapidly evaluate candidate genes and their interactions during heart development, which are crucial first steps toward deciphering oligogenic underpinnings of CHDs, including hypoplastic left hearts.
Collapse
Affiliation(s)
- Jeanne L Theis
- Cardiovascular Genetics Research LaboratoryRochesterUnited States
| | - Georg Vogler
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Maria A Missinato
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Tanja Nielsen
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
- Doctoral Degrees and Habilitations, Department of Biology, Chemistry, and Pharmacy, Freie Universität BerlinBerlinGermany
| | - Xin-Xin I Zeng
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | | | - Stanley M Walls
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Anaïs Kervadec
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - James N Kezos
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Katja Birker
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Megan M O'Byrne
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Zachary C Fogarty
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - André Terzic
- Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Center for Regenerative Medicine, Mayo ClinicRochesterUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Paul Grossfeld
- University of California San Diego, Rady’s HospitalSan DiegoUnited States
- Division of General Internal Medicine, Mayo ClinicRochesterUnited States
| | - Karen Ocorr
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Timothy J Nelson
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Center for Regenerative Medicine, Mayo ClinicRochesterUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Timothy M Olson
- Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Alexandre R Colas
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Rolf Bodmer
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| |
Collapse
|
15
|
Howard AM, Milner H, Hupp M, Willett C, Palermino K, Nowak SJ. Akirin is critical for early tinman induction and subsequent formation of the heart in Drosophila melanogaster. Dev Biol 2020; 469:1-11. [PMID: 32950464 DOI: 10.1016/j.ydbio.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 11/30/2022]
Abstract
The regulation of formation of the Drosophila heart by the Nkx 2.5 homologue Tinman is a key event during embryonic development. In this study, we identify the highly conserved transcription cofactor Akirin as a key factor in the earliest induction of tinman by the Twist transcription cofactor. akirin mutant embryos display a variety of morphological defects in the heart, including abnormal spacing between rows of aortic cells and abnormal patterning of the aortic outflow tract. akirin mutant embryos have a greatly reduced level of tinman transcripts, together with a reduction of Tinman protein in the earliest stages of cardiac patterning. Further, akirin mutants have reduced numbers of Tinman-positive cardiomyoblasts, concomitant with disrupted patterning and organization of the heart. Finally, despite the apparent formation of the heart in akirin mutants, these mutant hearts exhibit fewer coordinated contractions in akirin mutants compared with wild-type hearts. These results indicate that Akirin is crucial for the first induction of tinman by the Twist transcription factor, and that the success of the cardiac patterning program is highly dependent upon establishing the proper level of tinman at the earliest steps of the cardiac developmental pathway.
Collapse
Affiliation(s)
- Austin M Howard
- Master of Science in Integrative Biology Program, Kennesaw State University, USA; Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA
| | - Hayley Milner
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA
| | - Madison Hupp
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA
| | - Courtney Willett
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA
| | - Kristina Palermino
- Master of Science in Integrative Biology Program, Kennesaw State University, USA; Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA
| | - Scott J Nowak
- Master of Science in Integrative Biology Program, Kennesaw State University, USA; Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, 30144, USA.
| |
Collapse
|
16
|
Schroeder AM, Allahyari M, Vogler G, Missinato MA, Nielsen T, Yu MS, Theis JL, Larsen LA, Goyal P, Rosenfeld JA, Nelson TJ, Olson TM, Colas AR, Grossfeld P, Bodmer R. Model system identification of novel congenital heart disease gene candidates: focus on RPL13. Hum Mol Genet 2020; 28:3954-3969. [PMID: 31625562 DOI: 10.1093/hmg/ddz213] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/28/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
Genetics is a significant factor contributing to congenital heart disease (CHD), but our understanding of the genetic players and networks involved in CHD pathogenesis is limited. Here, we searched for de novo copy number variations (CNVs) in a cohort of 167 CHD patients to identify DNA segments containing potential pathogenic genes. Our search focused on new candidate disease genes within 19 deleted de novo CNVs, which did not cover known CHD genes. For this study, we developed an integrated high-throughput phenotypical platform to probe for defects in cardiogenesis and cardiac output in human induced pluripotent stem cell (iPSC)-derived multipotent cardiac progenitor (MCPs) cells and, in parallel, in the Drosophila in vivo heart model. Notably, knockdown (KD) in MCPs of RPL13, a ribosomal gene and SON, an RNA splicing cofactor, reduced proliferation and differentiation of cardiomyocytes, while increasing fibroblasts. In the fly, heart-specific RpL13 KD, predominantly at embryonic stages, resulted in a striking 'no heart' phenotype. KD of Son and Pdss2, among others, caused structural and functional defects, including reduced or abolished contractility, respectively. In summary, using a combination of human genetics and cardiac model systems, we identified new genes as candidates for causing human CHD, with particular emphasis on ribosomal genes, such as RPL13. This powerful, novel approach of combining cardiac phenotyping in human MCPs and in the in vivo Drosophila heart at high throughput will allow for testing large numbers of CHD candidates, based on patient genomic data, and for building upon existing genetic networks involved in heart development and disease.
Collapse
Affiliation(s)
- Analyne M Schroeder
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Massoud Allahyari
- Department of Pediatrics, UCSD School of Medicine, La Jolla, CA, USA
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Maria A Missinato
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Tanja Nielsen
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Michael S Yu
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jeanne L Theis
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Lars A Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Preeya Goyal
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Timothy J Nelson
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy M Olson
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexandre R Colas
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Paul Grossfeld
- Department of Pediatrics, UCSD School of Medicine, La Jolla, CA, USA
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
17
|
Daou R, Beißbarth T, Wingender E, Gültas M, Haubrock M. Constructing temporal regulatory cascades in the context of development and cell differentiation. PLoS One 2020; 15:e0231326. [PMID: 32275727 PMCID: PMC7147753 DOI: 10.1371/journal.pone.0231326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/20/2020] [Indexed: 12/02/2022] Open
Abstract
Cell differentiation is a complex process orchestrated by sets of regulators precisely appearing at certain time points, resulting in regulatory cascades that affect the expression of broader sets of genes, ending up in the formation of different tissues and organ parts. The identification of stage-specific master regulators and the mechanism by which they activate each other is a key to understanding and controlling differentiation, particularly in the fields of tissue regeneration and organoid engineering. Here we present a workflow that combines a comprehensive general regulatory network based on binding site predictions with user-provided temporal gene expression data, to generate a a temporally connected series of stage-specific regulatory networks, which we call a temporal regulatory cascade (TRC). A TRC identifies those regulators that are unique for each time point, resulting in a cascade that shows the emergence of these regulators and regulatory interactions across time. The model was implemented in the form of a user-friendly, visual web-tool, that requires no expert knowledge in programming or statistics, making it directly usable for life scientists. In addition to generating TRCs the tool links multiple interactive visual workflows, in which a user can track and investigate further different regulators, target genes, and interactions, directing the tool along the way into biologically sensible results based on the given dataset. We applied the TRC model on two different expression datasets, one based on experiments conducted on human induced pluripotent stem cells (hiPSCs) undergoing differentiation into mature cardiomyocytes and the other based on the differentiation of H1-derived human neuronal precursor cells. The model was successful in identifying previously known and new potential key regulators, in addition to the particular time points with which these regulators are associated, in cardiac and neural development.
Collapse
Affiliation(s)
- Rayan Daou
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Edgar Wingender
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Science, Georg-August University, Goettingen, Niedersachsen, Germany
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Goettingen, Niedersachsen, Germany
| | - Martin Haubrock
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goettingen, Niedersachsen, Germany
- * E-mail:
| |
Collapse
|
18
|
Ballinger- C, Anyagaligb O, Bernard J, Bierbower SM, Dupont-Ver EE, Ghoweri A, Greenhalgh A, Harrison D, Istas O, McNabb M, Saelinger C, Stanback A, Stanback M, Thibault O, Cooper RL. Effects of Bacterial Endotoxin (LPS) on Cardiac and Synaptic Function in Various Animal Models: Larval Drosophila, Crayfish, Crab and Rodent. ACTA ACUST UNITED AC 2019. [DOI: 10.3923/ijzr.2020.33.62] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
19
|
Potter S, Sifers J, Yocom E, Blümich SLE, Potter R, Nadolski J, Harrison DA, Cooper RL. Effects of inhibiting mTOR with rapamycin on behavior, development, neuromuscular physiology and cardiac function in larval Drosophila. Biol Open 2019; 8:bio.046508. [PMID: 31704693 PMCID: PMC6899040 DOI: 10.1242/bio.046508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rapamycin and other mTOR inhibitors are being heralded as possible treatments for many human ailments. It is currently being utilized clinically as an immunomodulator after transplantation procedures and as a treatment for certain forms of cancer, but it has numerous potential clinical indications. Some studies have shown profound effects on life cycle and muscle physiology, but these issues have not been addressed in an organism undergoing developmental processes. This paper fills this void by examining the effect of mTOR inhibition by rapamycin on several different qualities of larval Drosophila. Various dosages of the compound were fed to second instar larvae. These larvae were monitored for pupae formation to elucidate possible life cycle effects, and a delay to pupation was quantified. Behavioral deficits were documented in rapamycin-treated larvae. Electrophysiological measurements were taken to discern changes in muscle physiology and synaptic signaling (i.e. resting membrane potential, amplitude of excitatory post-synaptic potentials, synaptic facilitation). Pupation delay and effects on behavior that are likely due to synaptic alterations within the central nervous system were discovered in rapamycin-fed larvae. These results allow for several conclusions as to how mTOR inhibition by rapamycin affects a developing organism. This could eventually allow for a more informed decision when using rapamycin and other mTOR inhibitors to treat human diseases, especially in children and adolescents, to account for known side effects. Summary: Inhibiting mTOR by rapamycin delays pupation, reduced body wall contractions and mouth-hook movements while synaptic transmission appeared normal in larval Drosophila.
Collapse
Affiliation(s)
- Samuel Potter
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Jacob Sifers
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA.,Alice Lloyd College, 100 Purpose Road, Pippa Passes, KY, 41844, USA
| | - Emily Yocom
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA.,Kentucky Wesleyan College, Owensboro, KY, 42301, USA
| | - Sandra L E Blümich
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA.,Veterinärmedizinische Fakultät, Universität Leipzig, Leipzig, Germany
| | - Rachel Potter
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Jeremy Nadolski
- Department of Mathematical and Computational Sciences, Benedictine University, Lisle, IL, 60532 , USA
| | - Douglas A Harrison
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Robin L Cooper
- Deptartment of Biology and Center for Muscle Biology, University of Kentucky, Lexington, KY, 40506, USA
| |
Collapse
|
20
|
Ruan H, Liao Y, Ren Z, Mao L, Yao F, Yu P, Ye Y, Zhang Z, Li S, Xu H, Liu J, Diao L, Zhou B, Han L, Wang L. Single-cell reconstruction of differentiation trajectory reveals a critical role of ETS1 in human cardiac lineage commitment. BMC Biol 2019; 17:89. [PMID: 31722692 PMCID: PMC6854813 DOI: 10.1186/s12915-019-0709-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/15/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Cardiac differentiation from human pluripotent stem cells provides a unique opportunity to study human heart development in vitro and offers a potential cell source for cardiac regeneration. Compared to the large body of studies investigating cardiac maturation and cardiomyocyte subtype-specific induction, molecular events underlying cardiac lineage commitment from pluripotent stem cells at early stage remain poorly characterized. RESULTS In order to uncover key molecular events and regulators controlling cardiac lineage commitment from a pluripotent state during differentiation, we performed single-cell RNA-Seq sequencing and obtained high-quality data for 6879 cells collected from 6 stages during cardiac differentiation from human embryonic stem cells and identified multiple cell subpopulations with distinct molecular features. Through constructing developmental trajectory of cardiac differentiation and putative ligand-receptor interactions, we revealed crosstalk between cardiac progenitor cells and endoderm cells, which could potentially provide a cellular microenvironment supporting cardiac lineage commitment at day 5. In addition, computational analyses of single-cell RNA-Seq data unveiled ETS1 (ETS Proto-Oncogene 1) activation as an important downstream event induced by crosstalk between cardiac progenitor cells and endoderm cells. Consistent with the findings from single-cell analysis, chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-Seq) against ETS1 revealed genomic occupancy of ETS1 at cardiac structural genes at day 9 and day 14, whereas ETS1 depletion dramatically compromised cardiac differentiation. CONCLUSION Together, our study not only characterized the molecular features of different cell types and identified ETS1 as a crucial factor induced by cell-cell crosstalk contributing to cardiac lineage commitment from a pluripotent state, but may also have important implications for understanding human heart development at early embryonic stage, as well as directed manipulation of cardiac differentiation in regenerative medicine.
Collapse
Affiliation(s)
- Hang Ruan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA
| | - Yingnan Liao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Zongna Ren
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Lin Mao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Peng Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA
| | - Shengli Li
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA
| | - Hanshi Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Jiewei Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin St, MSB6.166, Houston, TX, 77030, USA.
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Beijing, 100037, People's Republic of China.
| |
Collapse
|
21
|
Kezos JN, Phillips MA, Thomas MD, Ewunkem AJ, Rutledge GA, Barter TT, Santos MA, Wong BD, Arnold KR, Humphrey LA, Yan A, Nouzille C, Sanchez I, Cabral LG, Bradley TJ, Mueller LD, Graves JL, Rose MR. Genomics of Early Cardiac Dysfunction and Mortality in Obese Drosophila melanogaster. Physiol Biochem Zool 2019; 92:591-611. [PMID: 31603376 DOI: 10.1086/706099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
In experimental evolution, we impose functional demands on laboratory populations of model organisms using selection. After enough generations of such selection, the resulting populations constitute excellent material for physiological research. An intense selection regime for increased starvation resistance was imposed on 10 large outbred Drosophila populations. We observed the selection responses of starvation and desiccation resistance, metabolic reserves, and heart robustness via electrical pacing. Furthermore, we sequenced the pooled genomes of these populations. As expected, significant increases in starvation resistance and lipid content were found in our 10 intensely selected SCO populations. The selection regime also improved desiccation resistance, water content, and glycogen content among these populations. Additionally, the average rate of cardiac arrests in our 10 obese SCO populations was double the rate of the 10 ancestral CO populations. Age-specific mortality rates were increased at early adult ages by selection. Genomic analysis revealed a large number of single nucleotide polymorphisms across the genome that changed in frequency as a result of selection. These genomic results were similar to those obtained in our laboratory from less direct selection procedures. The combination of extensive genomic and phenotypic differentiation between these 10 populations and their ancestors makes them a powerful system for the analysis of the physiological underpinnings of starvation resistance.
Collapse
|
22
|
Stanley CE, Mauss AS, Borst A, Cooper RL. The Effects of Chloride Flux on Drosophila Heart Rate. Methods Protoc 2019; 2:mps2030073. [PMID: 31443492 PMCID: PMC6789470 DOI: 10.3390/mps2030073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022] Open
Abstract
Approaches are sought after to regulate ionotropic and chronotropic properties of the mammalian heart. Electrodes are commonly used for rapidly exciting cardiac tissue and resetting abnormal pacing. With the advent of optogenetics and the use of tissue-specific expression of light-activated channels, cardiac cells cannot only be excited but also inhibited with ion-selective conductance. As a proof of concept for the ability to slow down cardiac pacing, anion-conducting channelrhodopsins (GtACR1/2) and the anion pump halorhodopsin (eNpHR) were expressed in hearts of larval Drosophila and activated by light. Unlike body wall muscles in most animals, the equilibrium potential for Cl− is more positive as compared to the resting membrane potential in larval Drosophila. As a consequence, upon activating the two forms of GtACR1 and 2 with low light intensity the heart rate increased, likely due to depolarization and opening of voltage-gated Ca2+ channels. However, with very intense light activation the heart rate ceases, which may be due to Cl– shunting to the reversal potential for chloride. Activating eNpHR hyperpolarizes body wall and cardiac muscle in larval Drosophila and rapidly decreases heart rate. The decrease in heart rate is related to light intensity. Intense light activation of eNpHR stops the heart from beating, whereas lower intensities slowed the rate. Even with upregulation of the heart rate with serotonin, the pacing of the heart was slowed with light. Thus, regulation of the heart rate in Drosophila can be accomplished by activating anion-conducting channelrhodopsins using light. These approaches are demonstrated in a genetically amenable insect model.
Collapse
Affiliation(s)
- Catherine E Stanley
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA
| | - Alex S Mauss
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Alexander Borst
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Robin L Cooper
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
23
|
Yang W, Yi K, Yu H, Ding Y, Li D, Wei Y, You T, Xie X. Correlation between pri-miR-124 (rs531564) polymorphism and congenital heart disease susceptibility in Chinese population at two different altitudes: a case-control and in silico study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:21983-21992. [PMID: 31144180 PMCID: PMC6657426 DOI: 10.1007/s11356-019-05350-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/01/2019] [Indexed: 06/09/2023]
Abstract
The development of congenital heart disease (CHD) is a complicated process and affected by multiple environmental factors, as genetic factors, and the interactions among those factors. Previous studies have shown that intrauterine hypoxic environment exposure is a risk factor of CHD, but the genetic factors involved in the process are not clear. In this study, given that tetralogy of Fallot (TOF) is a CHD with hypoxemia as its primary pathophysiological manifestation, an in silico analysis was performed to reveal the relationship between potential target genes (miR-124) with the energy metabolism in non-syndromic TOF patients' cardiomyocyte. Furthermore, the study investigated the correlation between the primary miR-124 (rs531564) polymorphism and CHD susceptibility in 432 sporadic patients and 450 controls from two different altitude provinces (city) in China. Our study indicated that the minor C allele of rs531564 correlated with reduced risk of CHD in the low altitude city. Besides, the C allele has elevated frequency in the high-altitude group. Therefore, our findings suggest that the minor C allele of rs531564 SNP may be involved in the reduction of the risk of CHD in a way that interacts with the intrauterine hypoxic environmental factors.
Collapse
Affiliation(s)
- Wenke Yang
- School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
- Gansu Cardiovascular Institute, People's Hospital of Lanzhou City, Lanzhou, 730050, China
| | - Kang Yi
- Department of Cardiac Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, China
| | - Hongmiao Yu
- School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - Yunhan Ding
- School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - Dehong Li
- Department of Cardiac Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yuping Wei
- Department of Cardiac Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Tao You
- Department of Cardiac Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China.
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, China.
| | - Xiaodong Xie
- School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China.
- Gansu Cardiovascular Institute, People's Hospital of Lanzhou City, Lanzhou, 730050, China.
| |
Collapse
|
24
|
Ninjurin1 regulates striated muscle growth and differentiation. PLoS One 2019; 14:e0216987. [PMID: 31091274 PMCID: PMC6519837 DOI: 10.1371/journal.pone.0216987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pressure overload due to aortic valve stenosis leads to pathological cardiac hypertrophy and heart failure. Hypertrophy is accompanied by an increase in myocyte surface area, which requires a proportional increase in the number of cell-cell and cell-matrix contacts to withstand enhanced workload. In a proteomic analysis we identified nerve injury-induced protein 1 (Ninjurin1), a 16kDa transmembrane cell-surface protein involved in cell adhesion and nerve repair, to be increased in hypertrophic hearts from patients with aortic stenosis. We hypothesised that Ninjurin1 is involved in myocyte hypertrophy. We analyzed cardiac biopsies from aortic-stenosis patients and control patients undergoing elective heart surgery. We studied cardiac hypertrophy in mice after transverse aortic constriction and angiotensin II infusions, and performed mechanistic analyses in cultured myocytes. We assessed the physiological role of ninjurin1 in zebrafish during heart and skeletal muscle development. Ninjurin1 was increased in hearts of aortic stenosis patients, compared to controls, as well as in hearts from mice with cardiac hypertrophy. Besides the 16kDa Ninjurin1 (Ninjurin1-16) we detected a 24kDa variant of Ninjurin1 (Ninjurin1-24), which was predominantly expressed during myocyte hypertrophy. We disclosed that the higher molecular weight of Ninjurin1-24 was caused by N-glycosylation. Ninjurin1-16 was contained in the cytoplasm of myocytes where it colocalized with stress-fibers. In contrast, Ninjurin1-24 was localized at myocyte membranes. Gain and loss-of-function experiments showed that Ninjurin1-24 plays a role in myocyte hypertrophy and myogenic differentiation in vitro. Reduced levels of ninjurin1 impaired cardiac and skeletal muscle development in zebrafish. We conclude that Ninjurin1 contributes to myocyte growth and differentiation, and that these effects are mainly mediated by N-glycosylated Ninjurin1-24.
Collapse
|
25
|
Anyagaligbo O, Bernard J, Greenhalgh A, Cooper RL. The effects of bacterial endotoxin (LPS) on cardiac function in a medicinal blow fly (Phaenicia sericata) and a fruit fly (Drosophila melanogaster). Comp Biochem Physiol C Toxicol Pharmacol 2019; 217:15-24. [PMID: 30448591 DOI: 10.1016/j.cbpc.2018.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022]
Abstract
The bacterial endotoxins, lipopolysaccharides (LPS), are known to have direct effects on mammalian heart cells; thus, LPS is likely to have some effects in other cardiac models. Drosophila melanogaster was used since it serves as a model for cardiac physiology. Larvae of blow flies (Phaenicia sericata) commonly used as therapy for debriding dead tissue, are exposed to high levels of bacterial endotoxins, but their mechanisms of LPS resistance are not entirely understood. Comparative effects of LPS on heart rate (HR) were examined for both Drosophila and blowfly larvae. Acute 10-min direct exposure of in situ heart tubes with saline containing 1, 100, and 500 μg/ml LPS from two common bacterial stains (Pseudomonas aeruginosa and Serratia marcescens) revealed a dose-dependent effect. The effects differed between the two fly models. Larval hearts of Drosophila stopped rapidly in low Ca2+ containing saline, but the hearts of blow flies appear unaffected for >30 min. S. marcescens increased HR initially in Drosophila followed by a reduction for low and high doses, but no change was observed in larvae of blow flies. Whereas P. aeruginosa at a high dose decreased HR in larvae of Drosophila but increased HR in larvae of blow flies. The goal of this study is to better the understanding in the direct action of LPS on HR. Knowing the acute and direct actions of LPS exposure on HR in different species of larvae may aid in understanding the underlying mechanisms in other animals during septicemia.
Collapse
Affiliation(s)
- Ogechi Anyagaligbo
- Department of Biology, Center for Muscle Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Jate Bernard
- Department of Biology, Center for Muscle Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Abigail Greenhalgh
- Department of Biology, Center for Muscle Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Robin L Cooper
- Department of Biology, Center for Muscle Biology, University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
26
|
Scimone ML, Wurtzel O, Malecek K, Fincher CT, Oderberg IM, Kravarik KM, Reddien PW. foxF-1 Controls Specification of Non-body Wall Muscle and Phagocytic Cells in Planarians. Curr Biol 2018; 28:3787-3801.e6. [PMID: 30471994 DOI: 10.1016/j.cub.2018.10.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/17/2018] [Accepted: 10/10/2018] [Indexed: 12/31/2022]
Abstract
Planarians are flatworms capable of regenerating any missing body part in a process requiring stem cells and positional information. Muscle is a major source of planarian positional information and consists of several types of fibers with distinct regulatory roles in regeneration. The transcriptional regulatory programs used to specify different muscle fibers are poorly characterized. Using single-cell RNA sequencing, we define the transcriptomes of planarian dorsal-ventral muscle (DVM), intestinal muscle (IM), and pharynx muscle. This analysis identifies foxF-1, which encodes a broadly conserved Fox-family transcription factor, as a master transcriptional regulator of all non-body wall muscle. The transcription factors encoded by nk4 and gata4/5/6-2 specify two different subsets of DVM, lateral and medial, respectively, whereas gata4/5/6-3 specifies IM. These muscle types all express planarian patterning genes. Both lateral and medial DVM are required for medial-lateral patterning in regeneration, whereas medial DVM and IM have a role in maintaining and regenerating intestine morphology. In addition to the role in muscle, foxF-1 is required for the specification of multiple cell types with transcriptome similarities, including high expression levels of cathepsin genes. These cells include pigment cells, glia, and several other cells with unknown function. cathepsin+ cells phagocytose E. coli, suggesting these are phagocytic cells. In conclusion, we describe a regulatory program for planarian muscle cell subsets and phagocytic cells, both driven by foxF-1. FoxF proteins specify different mesoderm-derived tissues in other organisms, suggesting that FoxF regulates formation of an ancient and broadly conserved subset of mesoderm derivatives in the Bilateria.
Collapse
Affiliation(s)
- M Lucila Scimone
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Omri Wurtzel
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Kathryn Malecek
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Christopher T Fincher
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Isaac M Oderberg
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Kellie M Kravarik
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Peter W Reddien
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA.
| |
Collapse
|
27
|
Zmojdzian M, de Joussineau S, Da Ponte JP, Jagla K. Distinct subsets of Eve-positive pericardial cells stabilise cardiac outflow and contribute to Hox gene-triggered heart morphogenesis in Drosophila. Development 2018; 145:dev.158717. [PMID: 29247145 PMCID: PMC5825839 DOI: 10.1242/dev.158717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/11/2017] [Indexed: 11/20/2022]
Abstract
The Drosophila heart, composed of discrete subsets of cardioblasts and pericardial cells, undergoes Hox-triggered anterior-posterior morphogenesis, leading to a functional subdivision into heart proper and aorta, with its most anterior part forming a funnel-shaped cardiac outflow. Cardioblasts differentiate into Tin-positive 'working myocytes' and Svp-expressing ostial cells. However, developmental fates and functions of heart-associated pericardial cells remain elusive. Here, we show that the pericardial cells that express the transcription factor Even Skipped adopt distinct fates along the anterior-posterior axis. Among them, the most anterior Antp-Ubx-AbdA-negative cells form a novel cardiac outflow component we call the outflow hanging structure, whereas the Antp-expressing cells differentiate into wing heart precursors. Interestingly, Hox gene expression in the Even Skipped-positive cells not only underlies their antero-posterior diversification, but also influences heart morphogenesis in a non-cell-autonomous way. In brief, we identify a new cardiac outflow component derived from a subset of Even Skipped-expressing cells that stabilises the anterior heart tip, and demonstrate non-cell-autonomous effects of Hox gene expression in the Even Skipped-positive cells on heart morphogenesis.
Collapse
Affiliation(s)
- Monika Zmojdzian
- GReD - INSERM U1103, CNRS UMR6293, University of Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Svetlana de Joussineau
- GReD - INSERM U1103, CNRS UMR6293, University of Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Jean Philippe Da Ponte
- GReD - INSERM U1103, CNRS UMR6293, University of Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Krzysztof Jagla
- GReD - INSERM U1103, CNRS UMR6293, University of Clermont Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
28
|
Gerke M, Bornberg-Bauer E, Jiang X, Fuellen G. Finding Common Protein Interaction Patterns Across Organisms. Evol Bioinform Online 2017. [DOI: 10.1177/117693430600200011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Protein interactions are an important resource to obtain an understanding of cell function. Recently, researchers have compared networks of interactions in order to understand network evolution. While current methods first infer homologs and then compare topologies, we here present a method which first searches for interesting topologies and then looks for homologs. PINA (protein interaction network analysis) takes the protein interaction networks of two organisms, scans both networks for subnetworks deemed interesting, and then tries to find orthologs among the interesting subnetworks. The application is very fast because orthology investigations are restricted to subnetworks like hubs and clusters that fulfill certain criteria regarding neighborhood and connectivity. Finally, the hubs or clusters found to be related can be visualized and analyzed according to protein annotation.
Collapse
Affiliation(s)
- Mirco Gerke
- Division of Bioinformatics, Biology Department, Schlossplatz 4, D-48149 Münster, Germany
- Institut für Informatik, Fachbereich Mathematik und Informatik, Einsteinstr. 62, D- 48149 Münster, Germany
| | - Erich Bornberg-Bauer
- Division of Bioinformatics, Biology Department, Schlossplatz 4, D-48149 Münster, Germany
| | - Xiaoyi Jiang
- Institut für Informatik, Fachbereich Mathematik und Informatik, Einsteinstr. 62, D- 48149 Münster, Germany
| | - Georg Fuellen
- Division of Bioinformatics, Biology Department, Schlossplatz 4, D-48149 Münster, Germany
- Department of Medicine, AG Bioinformatics, Domagkstr. 3, D-48149 Münster, Germany
| |
Collapse
|
29
|
Ahmad SM. Conserved signaling mechanisms in Drosophila heart development. Dev Dyn 2017; 246:641-656. [PMID: 28598558 PMCID: PMC11546222 DOI: 10.1002/dvdy.24530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/06/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Signal transduction through multiple distinct pathways regulates and orchestrates the numerous biological processes comprising heart development. This review outlines the roles of the FGFR, EGFR, Wnt, BMP, Notch, Hedgehog, Slit/Robo, and other signaling pathways during four sequential phases of Drosophila cardiogenesis-mesoderm migration, cardiac mesoderm establishment, differentiation of the cardiac mesoderm into distinct cardiac cell types, and morphogenesis of the heart and its lumen based on the proper positioning and cell shape changes of these differentiated cardiac cells-and illustrates how these same cardiogenic roles are conserved in vertebrates. Mechanisms bringing about the regulation and combinatorial integration of these diverse signaling pathways in Drosophila are also described. This synopsis of our present state of knowledge of conserved signaling pathways in Drosophila cardiogenesis and the means by which it was acquired should facilitate our understanding of and investigations into related processes in vertebrates. Developmental Dynamics 246:641-656, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shaad M. Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
30
|
Antagonistic BMP-cWNT signaling in the cnidarian Nematostella vectensis reveals insight into the evolution of mesoderm. Proc Natl Acad Sci U S A 2017; 114:E5608-E5615. [PMID: 28652368 DOI: 10.1073/pnas.1701607114] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Gastrulation was arguably the key evolutionary innovation that enabled metazoan diversification, leading to the formation of distinct germ layers and specialized tissues. Differential gene expression specifying cell fate is governed by the inputs of intracellular and/or extracellular signals. Beta-catenin/Tcf and the TGF-beta bone morphogenetic protein (BMP) provide critical molecular signaling inputs during germ layer specification in bilaterian metazoans, but there has been no direct experimental evidence for a specific role for BMP signaling during endomesoderm specification in the early branching metazoan Nematostella vectensis (an anthozoan cnidarian). Using forward transcriptomics, we show that beta-catenin/Tcf signaling and BMP2/4 signaling provide differential inputs into the cnidarian endomesodermal gene regulatory network (GRN) at the onset of gastrulation (24 h postfertilization) in N. vectensis Surprisingly, beta-catenin/Tcf signaling and BMP2/4 signaling regulate a subset of common downstream target genes in the GRN in opposite ways, leading to the spatial and temporal differentiation of fields of cells in the developing embryo. Thus, we show that regulatory interactions between beta-catenin/Tcf signaling and BMP2/4 signaling are required for the specification and determination of different embryonic regions and the patterning of the oral-aboral axis in Nematostella We also show functionally that the conserved "kernel" of the bilaterian heart mesoderm GRN is operational in N. vectensis, which reinforces the hypothesis that the endoderm and mesoderm in triploblastic bilaterians evolved from the bifunctional endomesoderm (gastrodermis) of a diploblastic ancestor, and that slow rhythmic contractions might have been one of the earliest functions of mesodermal tissue.
Collapse
|
31
|
Whole genome analysis of a schistosomiasis-transmitting freshwater snail. Nat Commun 2017; 8:15451. [PMID: 28508897 PMCID: PMC5440852 DOI: 10.1038/ncomms15451] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/30/2017] [Indexed: 12/31/2022] Open
Abstract
Biomphalaria snails are instrumental in transmission of the human blood fluke Schistosoma mansoni. With the World Health Organization's goal to eliminate schistosomiasis as a global health problem by 2025, there is now renewed emphasis on snail control. Here, we characterize the genome of Biomphalaria glabrata, a lophotrochozoan protostome, and provide timely and important information on snail biology. We describe aspects of phero-perception, stress responses, immune function and regulation of gene expression that support the persistence of B. glabrata in the field and may define this species as a suitable snail host for S. mansoni. We identify several potential targets for developing novel control measures aimed at reducing snail-mediated transmission of schistosomiasis.
Collapse
|
32
|
Kezos JN, Cabral LG, Wong BD, Khou BK, Oh A, Harb JF, Chiem D, Bradley TJ, Mueller LD, Rose MR. Starvation but not locomotion enhances heart robustness in Drosophila. JOURNAL OF INSECT PHYSIOLOGY 2017; 99:8-14. [PMID: 28285040 DOI: 10.1016/j.jinsphys.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 06/06/2023]
Abstract
Insects and vertebrates have multiple major physiological systems, each species having a circulatory system, a metabolic system, and a respiratory system that enable locomotion and survival in stressful environments, among other functions. Broadening our understanding of the physiology of Drosophila melanogaster requires the parsing of interrelationships among such major component physiological systems. By combining electrical pacing and flight exhaustion assays with manipulative conditioning, we have started to unpack the interrelationships between cardiac function, locomotor performance, and other functional characters such as starvation and desiccation resistance. Manipulative sequences incorporating these four physiological characters were applied to five D. melanogaster lab populations that share a common origin from the wild and a common history of experimental evolution. While exposure to starvation or desiccation significantly reduced flight duration, exhaustion due to flight only affected subsequent desiccation resistance. A strong association was found between flight duration and desiccation resistance, providing additional support for the hypothesis that these traits depend on glycogen and water content. However, there was negligible impact on rate of cardiac arrests from exhaustion by flight or exposure to desiccant. Brief periods of starvation significantly lowered the rate of cardiac arrest. These results provide suggestive support for the adverse impact of lipids on Drosophila heart robustness, a parallel result to those of many comparable studies in human cardiology. Overall, this study underscores clear distinctions among the connections between specific physiological responses to stress and specific types of physiological performance.
Collapse
Affiliation(s)
- James N Kezos
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Larry G Cabral
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Brandon D Wong
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Belinda K Khou
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Angela Oh
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Jerry F Harb
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Danny Chiem
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Timothy J Bradley
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Laurence D Mueller
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| | - Michael R Rose
- Department of Ecology and Evolutionary Biology, Francisco J. Ayala School of Biological Sciences, University of California, Irvine, CA 92697-2525, United States.
| |
Collapse
|
33
|
Burkhard S, van Eif V, Garric L, Christoffels VM, Bakkers J. On the Evolution of the Cardiac Pacemaker. J Cardiovasc Dev Dis 2017; 4:jcdd4020004. [PMID: 29367536 PMCID: PMC5715705 DOI: 10.3390/jcdd4020004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023] Open
Abstract
The rhythmic contraction of the heart is initiated and controlled by an intrinsic pacemaker system. Cardiac contractions commence at very early embryonic stages and coordination remains crucial for survival. The underlying molecular mechanisms of pacemaker cell development and function are still not fully understood. Heart form and function show high evolutionary conservation. Even in simple contractile cardiac tubes in primitive invertebrates, cardiac function is controlled by intrinsic, autonomous pacemaker cells. Understanding the evolutionary origin and development of cardiac pacemaker cells will help us outline the important pathways and factors involved. Key patterning factors, such as the homeodomain transcription factors Nkx2.5 and Shox2, and the LIM-homeodomain transcription factor Islet-1, components of the T-box (Tbx), and bone morphogenic protein (Bmp) families are well conserved. Here we compare the dominant pacemaking systems in various organisms with respect to the underlying molecular regulation. Comparative analysis of the pathways involved in patterning the pacemaker domain in an evolutionary context might help us outline a common fundamental pacemaker cell gene programme. Special focus is given to pacemaker development in zebrafish, an extensively used model for vertebrate development. Finally, we conclude with a summary of highly conserved key factors in pacemaker cell development and function.
Collapse
Affiliation(s)
- Silja Burkhard
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent van Eif
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Laurence Garric
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent M Christoffels
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
34
|
Tarifeño-Saldivia E, Lavergne A, Bernard A, Padamata K, Bergemann D, Voz ML, Manfroid I, Peers B. Transcriptome analysis of pancreatic cells across distant species highlights novel important regulator genes. BMC Biol 2017; 15:21. [PMID: 28327131 PMCID: PMC5360028 DOI: 10.1186/s12915-017-0362-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Defining the transcriptome and the genetic pathways of pancreatic cells is of great interest for elucidating the molecular attributes of pancreas disorders such as diabetes and cancer. As the function of the different pancreatic cell types has been maintained during vertebrate evolution, the comparison of their transcriptomes across distant vertebrate species is a means to pinpoint genes under strong evolutionary constraints due to their crucial function, which have therefore preserved their selective expression in these pancreatic cell types. RESULTS In this study, RNA-sequencing was performed on pancreatic alpha, beta, and delta endocrine cells as well as the acinar and ductal exocrine cells isolated from adult zebrafish transgenic lines. Comparison of these transcriptomes identified many novel markers, including transcription factors and signaling pathway components, specific for each cell type. By performing interspecies comparisons, we identified hundreds of genes with conserved enriched expression in endocrine and exocrine cells among human, mouse, and zebrafish. This list includes many genes known as crucial for pancreatic cell formation or function, but also pinpoints many factors whose pancreatic function is still unknown. A large set of endocrine-enriched genes can already be detected at early developmental stages as revealed by the transcriptomic profiling of embryonic endocrine cells, indicating a potential role in cell differentiation. The actual involvement of conserved endocrine genes in pancreatic cell differentiation was demonstrated in zebrafish for myt1b, whose invalidation leads to a reduction of alpha cells, and for cdx4, selectively expressed in endocrine delta cells and crucial for their specification. Intriguingly, comparison of the endocrine alpha and beta cell subtypes from human, mouse, and zebrafish reveals a much lower conservation of the transcriptomic signatures for these two endocrine cell subtypes compared to the signatures of pan-endocrine and exocrine cells. These data suggest that the identity of the alpha and beta cells relies on a few key factors, corroborating numerous examples of inter-conversion between these two endocrine cell subtypes. CONCLUSION This study highlights both evolutionary conserved and species-specific features that will help to unveil universal and fundamental regulatory pathways as well as pathways specific to human and laboratory animal models such as mouse and zebrafish.
Collapse
Affiliation(s)
- Estefania Tarifeño-Saldivia
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Arnaud Lavergne
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Alice Bernard
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Keerthana Padamata
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - David Bergemann
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Marianne L Voz
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Bernard Peers
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium.
| |
Collapse
|
35
|
Shahrestani P, Burke MK, Birse R, Kezos JN, Ocorr K, Mueller LD, Rose MR, Bodmer R. Experimental Evolution and Heart Function in Drosophila. Physiol Biochem Zool 2017; 90:281-293. [PMID: 28277957 DOI: 10.1086/689288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drosophila melanogaster is a good model species for the study of heart function. However, most previous work on D. melanogaster heart function has focused on the effects of large-effect genetic variants. We compare heart function among 18 D. melanogaster populations that have been selected for altered development time, aging, or stress resistance. We find that populations with faster development and faster aging have increased heart dysfunction, measured as percentage heart failure after electrical pacing. Experimental evolution of different triglyceride levels, by contrast, has little effect on heart function. Evolved differences in heart function correlate with allele frequency changes at many loci of small effect. Genomic analysis of these populations produces a list of candidate loci that might affect cardiac function at the intersection of development, aging, and metabolic control mechanisms.
Collapse
|
36
|
Abstract
The tumor suppressor Trp53 (p53) inhibits cell growth after acute stress by regulating gene transcription. The mammalian genome contains hundreds of p53-binding sites. However, whether p53 participates in the regulation of cardiac tissue homeostasis under normal conditions is not known. To examine the physiologic role of p53 in adult cardiomyocytes in vivo, Cre-loxP-mediated conditional gene targeting in adult mice was used. Genome-wide transcriptome analyses of conditional heart-specific p53 knockout mice were performed. Genome-wide annotation and pathway analyses of >5,000 differentially expressed transcripts identified many p53-regulated gene clusters. Correlative analyses identified >20 gene sets containing more than 1,000 genes relevant to cardiac architecture and function. These transcriptomic changes orchestrate cardiac architecture, excitation-contraction coupling, mitochondrial biogenesis, and oxidative phosphorylation capacity. Interestingly, the gene expression signature in p53-deficient hearts confers resistance to acute biomechanical stress. The data presented here demonstrate a role for p53, a previously unrecognized master regulator of the cardiac transcriptome. The complex contributions of p53 define a biological paradigm for the p53 regulator network in the heart under physiological conditions.
Collapse
|
37
|
Dadson K, Hauck L, Hao Z, Grothe D, Rao V, Mak TW, Billia F. The E3 ligase Mule protects the heart against oxidative stress and mitochondrial dysfunction through Myc-dependent inactivation of Pgc-1α and Pink1. Sci Rep 2017; 7:41490. [PMID: 28148912 PMCID: PMC5288653 DOI: 10.1038/srep41490] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2016] [Indexed: 01/03/2023] Open
Abstract
Cardiac homeostasis requires proper control of protein turnover. Protein degradation is principally controlled by the Ubiquitin-Proteasome System. Mule is an E3 ubiquitin ligase that regulates cellular growth, DNA repair and apoptosis to maintain normal tissue architecture. However, Mule’s function in the heart has yet to be described. In a screen, we found reduced Mule expression in left ventricular samples from end-stage heart failure patients. Consequently, we generated conditional cardiac-specific Mule knockout (Mule fl/fl(y);mcm) mice. Mule ablation in adult Mule fl/fl(y);mcm mice prevented myocardial c-Myc polyubiquitination, leading to c-Myc accumulation and subsequent reduced expression of Pgc-1α, Pink1, and mitochondrial complex proteins. Furthermore, these mice developed spontaneous cardiac hypertrophy, left ventricular dysfunction, and early mortality. Co-deletion of Mule and c-Myc rescued this phenotype. Our data supports an indispensable role for Mule in cardiac homeostasis through the regulation of mitochondrial function via maintenance of Pgc-1α and Pink1 expression and persistent negative regulation of c-Myc.
Collapse
Affiliation(s)
- Keith Dadson
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Ludger Hauck
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Zhenyue Hao
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Daniela Grothe
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Vivek Rao
- Division of Cardiovascular Surgery, UHN, Toronto, ON, M5G 2C4, Canada
| | - Tak W Mak
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Filio Billia
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada.,Division of Cardiology, University Health Network (UHN), Toronto, Ontario, Canada.,Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, M5G 1A8, Ontario Canada
| |
Collapse
|
38
|
Men J, Jerwick J, Wu P, Chen M, Alex A, Ma Y, Tanzi RE, Li A, Zhou C. Drosophila Preparation and Longitudinal Imaging of Heart Function In Vivo Using Optical Coherence Microscopy (OCM). J Vis Exp 2016. [PMID: 28060288 DOI: 10.3791/55002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Longitudinal study of the heartbeat in small animals contributes to understanding structural and functional changes during heart development. Optical coherence microscopy (OCM) has been demonstrated to be capable of imaging small animal hearts with high spatial resolution and ultrahigh imaging speed. The high image contrast and noninvasive properties make OCM ideal for performing longitudinal studies without requiring tissue dissections or staining. Drosophila has been widely used as a model organism in cardiac developmental studies due to its high number of orthologous human disease genes, its similarity of molecular mechanisms and genetic pathways with vertebrates, its short life cycle, and its low culture cost. Here, the experimental protocols are described for the preparation of Drosophila and optical imaging of the heartbeat with a custom OCM system throughout the life cycle of the specimen. By following the steps provided in this report, transverse M-mode and 3D OCM images can be acquired to conduct longitudinal studies of the Drosophila cardiac morphology and function. The en face and axial sectional OCM images and the heart rate (HR) and cardiac activity period (CAP) histograms, were also shown to analyze the heart structural changes and to quantify the heart dynamics during Drosophila metamorphosis, combined with the videos constructed with M-mode images to trace cardiac activity intuitively. Due to the genetic similarity between Drosophila and vertebrates, longitudinal study of heart morphology and dynamics in fruit flies could help reveal the origins of human heart diseases. The protocol here would provide an effective method to perform a wide range of studies to understand the mechanisms of cardiac diseases in humans.
Collapse
Affiliation(s)
- Jing Men
- Bioengineering Program, Lehigh University; Center for Photonics and Nanoelectronics, Lehigh University
| | - Jason Jerwick
- Center for Photonics and Nanoelectronics, Lehigh University; Department of Electrical and Computer Engineering, Lehigh University
| | - Penghe Wu
- Bioengineering Program, Lehigh University; Center for Photonics and Nanoelectronics, Lehigh University
| | - Mingming Chen
- Department of Electrical and Computer Engineering, Lehigh University; State Key Laboratory of Software Engineering, Wuhan University
| | - Aneesh Alex
- Center for Photonics and Nanoelectronics, Lehigh University; Department of Electrical and Computer Engineering, Lehigh University
| | - Yutao Ma
- State Key Laboratory of Software Engineering, Wuhan University
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School
| | - Airong Li
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School
| | - Chao Zhou
- Bioengineering Program, Lehigh University; Center for Photonics and Nanoelectronics, Lehigh University; Department of Electrical and Computer Engineering, Lehigh University;
| |
Collapse
|
39
|
Men J, Huang Y, Solanki J, Zeng X, Alex A, Jerwick J, Zhang Z, Tanzi RE, Li A, Zhou C. Optical Coherence Tomography for Brain Imaging and Developmental Biology. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2016; 22:6803213. [PMID: 27721647 PMCID: PMC5049888 DOI: 10.1109/jstqe.2015.2513667] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Optical coherence tomography (OCT) is a promising research tool for brain imaging and developmental biology. Serving as a three-dimensional optical biopsy technique, OCT provides volumetric reconstruction of brain tissues and embryonic structures with micrometer resolution and video rate imaging speed. Functional OCT enables label-free monitoring of hemodynamic and metabolic changes in the brain in vitro and in vivo in animal models. Due to its non-invasiveness nature, OCT enables longitudinal imaging of developing specimens in vivo without potential damage from surgical operation, tissue fixation and processing, and staining with exogenous contrast agents. In this paper, various OCT applications in brain imaging and developmental biology are reviewed, with a particular focus on imaging heart development. In addition, we report findings on the effects of a circadian gene (Clock) and high-fat-diet on heart development in Drosophila melanogaster. These findings contribute to our understanding of the fundamental mechanisms connecting circadian genes and obesity to heart development and cardiac diseases.
Collapse
Affiliation(s)
- Jing Men
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| | - Yongyang Huang
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| | - Jitendra Solanki
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| | - Xianxu Zeng
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
- Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China, 450000
| | - Aneesh Alex
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| | - Jason Jerwick
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| | - Zhan Zhang
- Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China, 450000
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA, 02129
| | - Airong Li
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA, 02129
| | - Chao Zhou
- Department of Electrical and Computer Engineering, Center for Photonics and Nanoelectronics, and Bioengineering Program, Lehigh University, Bethlehem, PA, USA, 18015
| |
Collapse
|
40
|
Lesch BJ, Silber SJ, McCarrey JR, Page DC. Parallel evolution of male germline epigenetic poising and somatic development in animals. Nat Genet 2016; 48:888-94. [PMID: 27294618 DOI: 10.1038/ng.3591] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
Abstract
Changes in gene regulation frequently underlie changes in morphology during evolution, and differences in chromatin state have been linked with changes in anatomical structure and gene expression across evolutionary time. Here we assess the relationship between evolution of chromatin state in germ cells and evolution of gene regulatory programs governing somatic development. We examined the poised (H3K4me3/H3K27me3 bivalent) epigenetic state in male germ cells from five mammalian and one avian species. We find that core genes poised in germ cells from multiple amniote species are ancient regulators of morphogenesis that sit at the top of transcriptional hierarchies controlling somatic tissue development, whereas genes that gain poising in germ cells from individual species act downstream of core poised genes during development in a species-specific fashion. We propose that critical regulators of animal development gained an epigenetically privileged state in germ cells, manifested in amniotes by H3K4me3/H3K27me3 poising, early in metazoan evolution.
Collapse
Affiliation(s)
| | - Sherman J Silber
- Infertility Center of St. Louis, St. Luke's Hospital, St. Louis, Missouri, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - David C Page
- Whitehead Institute, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Whitehead Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
41
|
Abstract
The high level of conservation of the cardiogenic gene regulatory network as well as of the cellular and physiological characteristics of the cardiomyocytes between fly and human, makes the small heart of this invertebrate the simplest and most flexible genetic system to dissect the fundamental molecular mechanisms that are brought into play during the development, the establishment and the maintenance of the cardiac function. The recent improvements in techniques of measurements of cardiac function made it possible to validate Drosophila as a model of cardiomyopathies and arrhythmias of genetic and metabolic origin or dependent of ageing. The heart of the fly thus represents a model of choice to identify genes and their interactions implicated in cardiac pathologies.
Collapse
Affiliation(s)
- Laurent Perrin
- Inserm UMR_S 1090, Technological advances for genomics and clinics (TAGC), parc scientifique de Luminy, case 908, 13288 Marseille Cedex 9, France - CNRS, Marseille, France - Aix-Marseille université, UMR_S 1090, TAGC, parc scientifique de Luminy, 13288 Marseille, France
| | - Laurence Röder
- Inserm UMR_S 1090, Technological advances for genomics and clinics (TAGC), parc scientifique de Luminy, case 908, 13288 Marseille Cedex 9, France - Aix-Marseille université, UMR_S 1090, TAGC, parc scientifique de Luminy, 13288 Marseille, France
| |
Collapse
|
42
|
Lovato TL, Cripps RM. Regulatory Networks that Direct the Development of Specialized Cell Types in the Drosophila Heart. J Cardiovasc Dev Dis 2016; 3. [PMID: 27695700 PMCID: PMC5044875 DOI: 10.3390/jcdd3020018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The Drosophila cardiac tube was once thought to be a simple linear structure, however research over the past 15 years has revealed significant cellular and molecular complexity to this organ. Prior reviews have focused upon the gene regulatory networks responsible for the specification of the cardiac field and the activation of cardiac muscle structural genes. Here we focus upon highlighting the existence, function, and development of unique cell types within the dorsal vessel, and discuss their correspondence to analogous structures in the vertebrate heart.
Collapse
|
43
|
Trujillo GV, Nodal DH, Lovato CV, Hendren JD, Helander LA, Lovato TL, Bodmer R, Cripps RM. The canonical Wingless signaling pathway is required but not sufficient for inflow tract formation in the Drosophila melanogaster heart. Dev Biol 2016; 413:16-25. [PMID: 26983369 PMCID: PMC4834244 DOI: 10.1016/j.ydbio.2016.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 10/22/2022]
Abstract
The inflow tracts of the embryonic Drosophila cardiac tube, termed ostia, arise in its posterior three segments from cardiac cells that co-express the homeotic transcription factor Abdominal-A (abdA), the orphan nuclear receptor Seven-up (Svp), and the signaling molecule Wingless (Wg). To define the roles of these factors in inflow tract development, we assessed their function in inflow tract formation. We demonstrate, using several criteria, that abdA, svp, and wg are each critical for normal inflow tract formation. We further show that Wg acts in an autocrine manner to impact ostia fate, and that it mediates this effect at least partially through the canonical Wg signaling pathway. By contrast, neither wg expression nor Wg signaling are sufficient for inflow tract formation when expressed in anterior Svp cells that do not normally form inflow tracts in the embryo. Instead, ectopic abd-A expression throughout the cardiac tube is required for the formation of ectopic inflow tracts, indicating that autocrine Wg signaling must be supplemented by additional Hox-dependent factors to effect inflow tract formation. Taken together, these studies define important cellular and molecular events that contribute to cardiac inflow tract development in Drosophila. Given the broad conservation of the cardiac regulatory network through evolution, our studies provide insight into mechanisms of cardiac development in higher animals.
Collapse
Affiliation(s)
- Gloriana V Trujillo
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA; Sanford Burnham Medical Research Institute, Development and Aging Program, La Jolla, CA 92037, USA
| | - Dalea H Nodal
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Candice V Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jill D Hendren
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Lynda A Helander
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - TyAnna L Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Rolf Bodmer
- Sanford Burnham Medical Research Institute, Development and Aging Program, La Jolla, CA 92037, USA
| | - Richard M Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
44
|
Taghli-Lamallem O, Plantié E, Jagla K. Drosophila in the Heart of Understanding Cardiac Diseases: Modeling Channelopathies and Cardiomyopathies in the Fruitfly. J Cardiovasc Dev Dis 2016; 3:jcdd3010007. [PMID: 29367558 PMCID: PMC5715700 DOI: 10.3390/jcdd3010007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 01/23/2016] [Accepted: 02/06/2016] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases and, among them, channelopathies and cardiomyopathies are a major cause of death worldwide. The molecular and genetic defects underlying these cardiac disorders are complex, leading to a large range of structural and functional heart phenotypes. Identification of molecular and functional mechanisms disrupted by mutations causing channelopathies and cardiomyopathies is essential to understanding the link between an altered gene and clinical phenotype. The development of animal models has been proven to be efficient for functional studies in channelopathies and cardiomyopathies. In particular, the Drosophila model has been largely applied for deciphering the molecular and cellular pathways affected in these inherited cardiac disorders and for identifying their genetic modifiers. Here we review the utility and the main contributions of the fruitfly models for the better understanding of channelopathies and cardiomyopathies. We also discuss the investigated pathological mechanisms and the discoveries of evolutionarily conserved pathways which reinforce the value of Drosophila in modeling human cardiac diseases.
Collapse
Affiliation(s)
- Ouarda Taghli-Lamallem
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France.
| | - Emilie Plantié
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France.
| | - Krzysztof Jagla
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
45
|
Asadzadeh J, Neligan N, Kramer SG, Labrador JP. Tinman Regulates NetrinB in the Cardioblasts of the Drosophila Dorsal Vessel. PLoS One 2016; 11:e0148526. [PMID: 26840059 PMCID: PMC4740434 DOI: 10.1371/journal.pone.0148526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/05/2016] [Indexed: 11/18/2022] Open
Abstract
Morphogenesis of the Drosophila dorsal vessel (DV) shares similarities with that of the vertebrate heart. Precursors line up at both sides of the embryo, migrate towards the midline and fuse to form a tubular structure. Guidance receptors and their ligands have been implicated in this process in vertebrates and invertebrates, as have been a series of evolutionarily conserved cardiogenic transcriptional regulators including Tinman, the Drosophila homolog of the transcription factor Nkx-2.5. NetrinB (NetB), a repulsive ligand for the Unc-5 receptor is required to preserve the dorsal vessel hollow. It localizes to the luminal space of the dorsal vessel but its source and its regulation is unknown. Here, using genetics together with in situ hybridization with single cell resolution, we show how tin is required for NetrinB expression in cardioblasts during DV tubulogenesis and sufficient to promote NetB transcription ectopically. We further identify a dorsal vessel-specific NetB enhancer and show that it is also regulated by tin in a similar fashion to NetB.
Collapse
Affiliation(s)
- Jamshid Asadzadeh
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Niamh Neligan
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Sunita G. Kramer
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Juan-Pablo Labrador
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
46
|
Ekhteraei-Tousi S, Mohammad-Soltani B, Sadeghizadeh M, Mowla SJ, Parsi S, Soleimani M. Inhibitory effect of hsa-miR-590-5p on cardiosphere-derived stem cells differentiation through downregulation of TGFB signaling. J Cell Biochem 2016; 116:179-91. [PMID: 25163461 DOI: 10.1002/jcb.24957] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 08/22/2014] [Indexed: 11/08/2022]
Abstract
The cardiac cells generation via stem cells differentiation is a promising approach to restore the myocardial infarction. Promoted by our primary bioinformatics analysis as well as some previously published data on potential role of hsa-miR-590-3p in cardiogenesis, we have tried to decipher the role of miR-590-5p during the course of differentiation of cardiosphere-derived cells (CDCs). The differentiation induction of CDCs by TGFB1 was confirmed by real-time PCR, ICC, and flow cytometry. The expression pattern of hsa-miR-590-5p and some related genes were examined during the differentiation process. In order to study the role of miR-590-5p in cardiac differentiation, the effect of miR-590 overexpression in CDCs was studied. Evaluating the expression patterns of miR-590 and its potential targets (TGFBRs) during the course of differentiation, demonstrated a significant downregulation of miR-590 and an upregulation of TGFBR2, following the treatment of CDCs with TGFB1. Therefore, we proposed a model in which TGFB1 exerts its differentiation induction via downregulation of miR-590, and hence the higher transcriptional expression level of TGFBR2. In accordance with our proposed model, transfection of CDCs by a pLenti-III-hsa-mir-590-GFP expression vector before or after the first TGFB1 treatment caused a significant alteration in the expression levels of TGFBRs. Moreover, our data revealed that overexpression of miR-590 before TGFB1 induction was able to attenuate the CDCs differentiation probably via the reduction of TGFBR2 expression level. Altogether, our data suggest an inhibitory role of miR-590 during the cardiac differentiation of CDCs which its suppression might elevate the rate of differentiation.
Collapse
Affiliation(s)
- Samaneh Ekhteraei-Tousi
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
47
|
Ahmad SM, Bhattacharyya P, Jeffries N, Gisselbrecht SS, Michelson AM. Two Forkhead transcription factors regulate cardiac progenitor specification by controlling the expression of receptors of the fibroblast growth factor and Wnt signaling pathways. Development 2015; 143:306-17. [PMID: 26657774 PMCID: PMC4725337 DOI: 10.1242/dev.122952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 11/26/2015] [Indexed: 11/20/2022]
Abstract
Cardiogenesis involves the coordinated regulation of multiple biological processes by a finite set of transcription factors (TFs). Here, we show that the Forkhead TFs Checkpoint suppressor homologue (CHES-1-like) and Jumeau (Jumu), which govern cardiac progenitor cell divisions by regulating Polo kinase activity, play an additional, mutually redundant role in specifying the cardiac mesoderm (CM) as eliminating the functions of both Forkhead genes in the same Drosophila embryo results in defective hearts with missing hemisegments. This process is mediated by the Forkhead TFs regulating the fibroblast growth factor receptor Heartless (Htl) and the Wnt receptor Frizzled (Fz): CHES-1-like and jumu exhibit synergistic genetic interactions with htl and fz in CM specification, thereby implying that they function through the same genetic pathways, and transcriptionally activate the expression of both receptor-encoding genes. Furthermore, ectopic overexpression of either htl or fz in the mesoderm partially rescues the defective CM specification phenotype in embryos lacking both Forkhead genes. Together, these data emphasize the functional redundancy that leads to robustness in the cardiac progenitor specification process, and illustrate the pleiotropic functions of Forkhead TFs in different aspects of cardiogenesis. Summary: Checkpoint suppressor homologue and Jumeau, which are known to govern cardiac progenitor cell divisions, play additional, mutually redundant roles in specifying cardiac mesoderm in Drosophila.
Collapse
Affiliation(s)
- Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pritha Bhattacharyya
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen S Gisselbrecht
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan M Michelson
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Asadzadeh J, Neligan N, Canabal-Alvear JJ, Daly AC, Kramer SG, Labrador JP. The Unc-5 Receptor Is Directly Regulated by Tinman in the Developing Drosophila Dorsal Vessel. PLoS One 2015; 10:e0137688. [PMID: 26356221 PMCID: PMC4565662 DOI: 10.1371/journal.pone.0137688] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 08/19/2015] [Indexed: 01/05/2023] Open
Abstract
During early heart morphogenesis cardiac cells migrate in two bilateral opposing rows, meet at the dorsal midline and fuse to form a hollow tube known as the primary heart field in vertebrates or dorsal vessel (DV) in Drosophila. Guidance receptors are thought to mediate this evolutionarily conserved process. A core of transcription factors from the NK2, GATA and T-box families are also believed to orchestrate this process in both vertebrates and invertebrates. Nevertheless, whether they accomplish their function, at least in part, through direct or indirect transcriptional regulation of guidance receptors is currently unknown. In our work, we demonstrate how Tinman (Tin), the Drosophila homolog of the Nkx-2.5 transcription factor, regulates the Unc-5 receptor during DV tube morphogenesis. We use genetics, expression analysis with single cell mRNA resolution and enhancer-reporter assays in vitro or in vivo to demonstrate that Tin is required for Unc-5 receptor expression specifically in cardioblasts. We show that Tin can bind to evolutionary conserved sites within an Unc-5 DV enhancer and that these sites are required for Tin-dependent transactivation both in vitro and in vivo.
Collapse
Affiliation(s)
- Jamshid Asadzadeh
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Niamh Neligan
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Judith J. Canabal-Alvear
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Amanda C. Daly
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sunita Gupta Kramer
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Juan-Pablo Labrador
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
49
|
Lovato TL, Sensibaugh CA, Swingle KL, Martinez MM, Cripps RM. The Drosophila Transcription Factors Tinman and Pannier Activate and Collaborate with Myocyte Enhancer Factor-2 to Promote Heart Cell Fate. PLoS One 2015. [PMID: 26225919 PMCID: PMC4520567 DOI: 10.1371/journal.pone.0132965] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Expression of the MADS domain transcription factor Myocyte Enhancer Factor 2 (MEF2) is regulated by numerous and overlapping enhancers which tightly control its transcription in the mesoderm. To understand how Mef2 expression is controlled in the heart, we identified a late stage Mef2 cardiac enhancer that is active in all heart cells beginning at stage 14 of embryonic development. This enhancer is regulated by the NK-homeodomain transcription factor Tinman, and the GATA transcription factor Pannier through both direct and indirect interactions with the enhancer. Since Tinman, Pannier and MEF2 are evolutionarily conserved from Drosophila to vertebrates, and since their vertebrate homologs can convert mouse fibroblast cells to cardiomyocytes in different activator cocktails, we tested whether over-expression of these three factors in vivo could ectopically activate known cardiac marker genes. We found that mesodermal over-expression of Tinman and Pannier resulted in approximately 20% of embryos with ectopic Hand and Sulphonylurea receptor (Sur) expression. By adding MEF2 alongside Tinman and Pannier, a dramatic expansion in the expression of Hand and Sur was observed in almost all embryos analyzed. Two additional cardiac markers were also expanded in their expression. Our results demonstrate the ability to initiate ectopic cardiac fate in vivo by the combination of only three members of the conserved Drosophila cardiac transcription network, and provide an opportunity for this genetic model system to be used to dissect the mechanisms of cardiac specification.
Collapse
Affiliation(s)
- TyAnna L. Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Cheryl A. Sensibaugh
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Kirstie L. Swingle
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Melody M. Martinez
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Richard M. Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
- * E-mail:
| |
Collapse
|
50
|
Rebeiz M, Patel NH, Hinman VF. Unraveling the Tangled Skein: The Evolution of Transcriptional Regulatory Networks in Development. Annu Rev Genomics Hum Genet 2015; 16:103-31. [PMID: 26079281 DOI: 10.1146/annurev-genom-091212-153423] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The molecular and genetic basis for the evolution of anatomical diversity is a major question that has inspired evolutionary and developmental biologists for decades. Because morphology takes form during development, a true comprehension of how anatomical structures evolve requires an understanding of the evolutionary events that alter developmental genetic programs. Vast gene regulatory networks (GRNs) that connect transcription factors to their target regulatory sequences control gene expression in time and space and therefore determine the tissue-specific genetic programs that shape morphological structures. In recent years, many new examples have greatly advanced our understanding of the genetic alterations that modify GRNs to generate newly evolved morphologies. Here, we review several aspects of GRN evolution, including their deep preservation, their mechanisms of alteration, and how they originate to generate novel developmental programs.
Collapse
Affiliation(s)
- Mark Rebeiz
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260;
| | | | | |
Collapse
|